Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Aging (Albany NY) ; 16(2): 1897-1910, 2024 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-38271139

RESUMO

The expression level of RNA-binding proteins (RBPs) is dysregulated in oral squamous cell carcinoma (OSCC) and other types of cancer. Among the RBPs, IMP3 is involved in the progression of OSCC. However, the regulation of mRNA fate by IMP3 in OSCC remains less understood. We analyzed the expression level of IMP3 and E2F5 in OSCC tissues and cell lines by immunohistochemistry, qRT-PCR and Western blot. Subsequently, to further investigate the effect of IMP3 on E2F5 expression, we used siRNAs to silence IMP3 expression in OSCC cell lines SCC-25 and SCC-4. The binding site of E2F5 mRNA and IMP3 was confirmed by RNA immunoprecipitation (RIP). Finally, the function of IMP3 and E2F5 was investigated in viro and in xenograft mouse models. Here we report a positive correlation between IMP3 and E2F5 expression in OSCC, which are involved in cell proliferation and cell cycle. Mechanistically, E2F5 mRNA is bound by IMP3 protein, and silencing it leads to a shortened mRNA half-life and reduced protein expression. Also, knockdown of IMP3 inhibited allograft tumor progression in vivo. These studies reveal the molecular mechanism by which IMP3 regulates E2F5 mRNA stability and identify IMP3/E2F5 as a potential therapeutic target in OSCC.


Assuntos
Fator de Transcrição E2F5 , MicroRNAs , Neoplasias Bucais , Proteínas de Ligação a RNA , Carcinoma de Células Escamosas de Cabeça e Pescoço , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células/genética , Fator de Transcrição E2F5/genética , Fator de Transcrição E2F5/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo
2.
Dig Dis Sci ; 69(2): 463-475, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38087129

RESUMO

BACKGROUND: Gallbladder cancer (GBC) remains a serious cause of cancer-related mortality across the globe. E2F5 has been identified to as a known oncogene in various cancers. However, the special functions of E2F5 have not been investigated in GBC. AIMS: To explore the regulatory functions of E2F5 and its related molecular regulatory mechanism in GBC progression. METHODS: The expression of genes were examined through qRT-PCR, western blot and IHC assay. The cell proliferation was assessed through CCK-8 and EDU assays. The cytotoxicity was tested through LDH assay. The percentage of CD8+ T cells and cell apoptosis were evaluated through flow cytometry. The binding ability was detected through luciferase reporter assay. The tumor growth was assessed through in vivo assays. RESULTS: In this study, it was demonstrated that E2F5 expression was evaluated in GBC, and resulted into poor prognosis. Bioinformatics analysis revealed E2F5 as a target for let-7d-5p, which when overexpressed, suppressed the metastasis and proliferation of GBC through the downregulation of E2F5. It was discovered that E2F5 activates JAK2/STAT3 signaling which is suppressed by let-7d-5p, implicating this pathway as one of the effectors of the oncogenic effects of ESF5 in GBC. E2F5 had been confirmed to aggravate tumor growth in vivo. CONCLUSION: E2F5 targeted by let-7d-5p facilitated cell proliferation, metastasis and immune escape in GBC through the JAK2/STAT3 pathway.


Assuntos
Carcinoma in Situ , Neoplasias da Vesícula Biliar , MicroRNAs , Humanos , Neoplasias da Vesícula Biliar/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Fator de Transcrição E2F5/genética , Fator de Transcrição E2F5/metabolismo
3.
Cell Signal ; 112: 110923, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37827344

RESUMO

Hepatocellular carcinoma (HCC) is one of the most aggressive human malignancies worldwide. However, the molecular mechanism of HCC metastasis is largely unknown. Long non-coding RNA (lncRNA) plays a key role in gene regulation, and dysregulation of lncRNA is critical to cancer metastasis. LINC01980 has been reported in ESCC recently, but the mechanism underlying its function in HCC is still unknown. In this study, we found that LINC01980 was upregulated and associated with notably poor overall survival in HCC patients. Functionally, LINC01980 played a carcinogenic role and promoted HCC metastasis. Mechanically, LINC01980 enhanced the E2F5 expression via competitively binding miR-376b-5p, thereby inducing epithelial-mesenchymal transition and promoting HCC cells migration and invasion. In addition, LINC01980-mediated HCC cells metastasis was dependent on E2F5. What's more, TGF-ß activated LINC01980 transcription through the canonical TGF-ß/SMAD signaling pathway in HCC. In conclusion, LINC01980, activated by the canonical TGF-ß/SMAD pathway, promoted HCC metastasis via miR-376b-5p/E2F5 axis. Therefore, LINC01980 might be a potential prognostic biomarker and therapeutic target of HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Movimento Celular/genética , Fator de Transcrição E2F5/genética , Fator de Transcrição E2F5/metabolismo
4.
Zhonghua Nan Ke Xue ; 29(2): 113-119, 2023 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-37847082

RESUMO

OBJECTIVE: To analyze the effects of lncRNA SNHG12 on the proliferation, migration and invasiveness of PCa cells by regulating the expression of E2F5. METHODS: Using real time fluorescence RT-PCR, we detected the expressions of lncRNA SNHG12 and E2F5, constructed the PC3 cells inhibiting the lncRNA SNHG12 expression. After transfection of the PC3 cells, we divided them into an NC, a si-NC, a si-SNHG12, a si-E2F5, a si-SNHG12+OE-si-NC, and a si-SNHG12+OE-E2F5 group, followed by examination of the proliferation, apoptosis, migration and invasiveness of the cells in different groups. RESULTS: The expressions of lncRNA SNHG12 and E2F5 were significantly up-regulated in the PCa tissue compared with those in the adjacent tissue (P < 0.05), remarkably higher in the DU145, LNCaP and PC3 groups than in the RWPE-1 group, the highest in the PC3 group (P < 0.05). The expression of SNHG12 was markedly down-regulated in the si-SNHG12 group (P < 0.05) in comparison with that in the si-NC group, indicating the successful construction of a PC3 cell line interfering with the lncRNA SNHG12 expression. Compared with the si-NC group, the si-SNHG12 group showed significant decreases in the values of CyclinD1, MMP-9 and OD and the numbers of migrating and invading cells, and an increase in apoptotic cells (P < 0.05), while the si-E2F5 group exhibited a remarkably down-regulated expression of E2F5 (P < 0.05), reduced values of CyclinD1, MMP-9 and OD, decreased numbers of migrating and invading cells and an increased number of apoptotic cells (P < 0.05). The dual luciferase report test showed that E2F5 reduced the luciferase activity of SNHG12 (P < 0.05 and had an insignificant impact on the luciferase activity of MUT-SNHG12 (P > 0.05). Inhibiting the expression of lncRNA SNHG12 resulted in significant decreases in the expression of E2F5, values of CyclinD1, MMP-9 and OD and numbers of migrating and invading cells, but an increase in apoptotic cells (P < 0.05). The E2F5 expression, the CyclinD1, MMP-9 and OD values and the numbers of migrating and invading cells were markedly increased while the number of apoptotic cells decreased in the si-SNHG12+OE-E2F5 group compared with those in the si-SNHG12+OE-si-NC group (P < 0.05). CONCLUSION: Interfering with the expression of lncRNA SNHG12 can regulate that of E2F5, inhibit the proliferation, migration and invasiveness of PCa cells and promote their apoptosis.


Assuntos
MicroRNAs , Neoplasias da Próstata , RNA Longo não Codificante , Masculino , Humanos , RNA Longo não Codificante/genética , Metaloproteinase 9 da Matriz/genética , Movimento Celular/genética , Proliferação de Células , Linhagem Celular Tumoral , Neoplasias da Próstata/genética , Apoptose/genética , Regulação Neoplásica da Expressão Gênica , Luciferases/genética , MicroRNAs/genética , Fator de Transcrição E2F5/genética
5.
Biomed Res Int ; 2022: 6918396, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35909488

RESUMO

Background: Retinoblastoma (RB) is the commonest primary intraocular malignancy during childhood. Circular RNAs (circRNAs) act as regulators in RB development, and hsa_circ_E2F5 (circ_0084811 in this study) was found to be highly expressed in RB cells, so we wanted to identify its detailed molecular mechanism. Methods: The expression level of circ_0084811 in RB cells was tested by RT-qPCR and its effects on RB cells were evaluated through functional assays. The regulatory mechanism that circ_0084811 may exert in RB progression was testified through mechanism experiments. Results: High circ_0084811 expression in RB cells facilitated cell proliferation but inhibited cell apoptosis. The enrichment of acetylation of histone 3 lysine 27 (H3K27ac) in circ_0084811 promoter induced circ_0084811 upregulation. Moreover, circ_0084811 regulated E2F transcription factor 5 (E2F5) expression via sponging microRNA-18a-5p (miR-18a-5p) and microRNA-18b-5p (miR-18b-5p). Conclusion: circ_0084811 modulated RB progression via the miR-18a-5p/miR-18b-5p/E2F5 axis.


Assuntos
MicroRNAs , Neoplasias da Retina , Retinoblastoma , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/genética , Fator de Transcrição E2F5 , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Retina/genética , Retinoblastoma/genética
6.
Cell Signal ; 99: 110440, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35985439

RESUMO

BACKGROUND: Pancreatic cancer belongs to lethal cancer with limited efficient treatment currently, and its main cause of death is rapid tumor growth and early metastasis. N6-methyladenosine (m6A) modification is a new method of epigenetic gene regulation involved in tumor progression, in which methyltransferase-like 3(METTL3) is the sole catalytic subunit. However, the role of METTL3 in pancreatic cancer remains to be explored. METHODS: m6A level was measured using MeRIP assay, and RT-qPCR and western blot were applied to determine mRNA and protein expression, respectively. Cellular behaviors were detected using CCK-8, EdU, wound healing and transwell assays. Xenograft assays were conducted to further verify the roles of METTL3 in pancreatic cancer. RESULTS: METTL3 was highly expressed in pancreatic cancer. However, downregulation of METTL3 restrained the viability, migration and invasion of pancreatic cancer cells. Moreover, E2F5 was found to be positively regulated by METTL3. Intriguingly, the anti-tumor functions of METTL3 knockdown in the phenotype of pancreatic cancer cells were overturned by overexpression of E2F5. Silencing METTL3 resulted in the decreased stability of E2F5 by methylating E2F5. CONCLUSIONS: In conclusion, METTL3 can promote the malignant progression of pancreatic cancer by modifying E2F5 through m6A methylation to promote its stability.


Assuntos
Neoplasias Pancreáticas , Sincalida , Adenosina/metabolismo , Fator de Transcrição E2F5 , Humanos , Metiltransferases/genética , Metiltransferases/metabolismo , Neoplasias Pancreáticas/genética , RNA Mensageiro/metabolismo , Neoplasias Pancreáticas
7.
Dig Liver Dis ; 54(7): 937-945, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34583905

RESUMO

The underlying mechanisms of E2F5 upregulation and its pro-tumor functions have not been elucidated in gastric cancer (GC). Here, the expression, prognostic value, mutation status, and promoter methylation of E2F5 were evaluated. The effects of E2F5 depletion on cell proliferation and invasion in GC, were also assessed through in vitro experiments. Additionally, gene set enrichment analysis (GSEA) was applied to analyze the potential downstream regulator of E2F5. The study also assessed the correlation and transcription regulation between E2F5 and UBE2T. Finally, the roles of UBE2T in E2F5-related pro-tumor functions were examined. The findings revealed that E2F5 was upregulated and showed remarkable association with pathological variables and prognosis. Hypomethylation of the E2F5 promoter predicted poor prognosis and partially caused E2F5 upregulation in GC. E2F5 knockdown significantly inhibited the proliferation and invasion of GC cells. E2F5 had a significant positive correlation with UBE2T in GC. Mechanistically, E2F5 promoted UBE2T transcription and UBE2T overexpression reversed the effects of E2F5 depletion on the proliferation and invasion of cells in GC. Taken together, this study originally confirmed the upregulation of E2F5 in GC, revealed that E2F5 can directly upregulate UBE2T transcription, and subsequently promote the malignant progression, which highlights that the E2F5/UBE2T axis can potentially be used in the diagnosis and treatment of GC.


Assuntos
Fator de Transcrição E2F5/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Gástricas , Enzimas de Conjugação de Ubiquitina/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Humanos , Prognóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Regulação para Cima
8.
Bosn J Basic Med Sci ; 22(2): 205-216, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-34784267

RESUMO

Circular RNA (circRNA) is a key regulator of tumor progression. However, the role of circFOXM1 in glioblastoma (GBM) progression is unclear. The aim of this study was to investigate the role of circFOXM1 in GBM progression. The expression levels of circFOXM1, miR-577, and E2F transcription factor 5 (E2F5) were examined by real-time quantitative polymerase chain reaction. Cell counting kit 8 assay, EdU staining, and transwell assay were used to detect cell proliferation, migration, and invasion. The levels of glutamine, glutamate, and α-ketoglutarate were determined to evaluate the glutaminolysis ability of cells. Protein expression was tested by Western blot analysis. Dual-luciferase reporter assay, RNA pull-down assay, and RNA immunoprecipitation assay were employed to verify the interaction between miR-577 and circFOXM1 or E2F5. Mice xenograft model for GBM was constructed to perform in vivo experiments. Our results showed that circFOXM1 was highly expressed in GBM tumor tissues and cells. Silencing of cir FOXM1 inhibited GBM cell proliferation, migration, invasion, glutaminolysis, as well as tumor growth. MiR-577 could be sponged by circFOXM1, and its inhibitor could reverse the suppressive effect of circFOXM1 downregulation on GBM progression. E2F5 was a target of miR-577, and the effect of its knockdown on GBM progression was consistent with that of circFOXM1 silencing. CircFOXM1 positively regulated E2F5 expression, while miR-577 negatively regulated E2F5 expression. In conclusion, our data confirmed that circFOXM1 could serve as a sponge of miR-577 to enhance the progression of GBM by targeting E2F5, which revealed that circFOXM1 might be a biomarker for GBM treatment.


Assuntos
Fator de Transcrição E2F5 , Glioblastoma , MicroRNAs , RNA Circular , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células , Fator de Transcrição E2F5/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Xenoenxertos , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , RNA Circular/metabolismo , Transdução de Sinais
9.
Anticancer Res ; 41(3): 1315-1325, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33788723

RESUMO

BACKGROUND/AIM: The aim of this study was to reveal the novel roles of calmodulin 2 (CALM2) in hepatocellular carcinoma (HCC) progression. MATERIALS AND METHODS: The effects of knockdown of CALM2 expression by siRNA were investigated using various experimental approaches in both cellular and molecular levels. RESULTS: Silencing of CALM2 inhibited HCC cell proliferation and colony formation through induction of apoptosis. At the molecular level, CALM2-specific knockdown led to the common dysregulation of 154 genes in HCC cells. Notably, E2F transcription factor 5 (E2F5), which is functionally associated with migration, invasion and proliferation, was generally down-regulated. These functional associations were confirmed in HCC clinical samples. Reflecting the molecular changes, CALM2 knockdown reduced the migration and invasion abilities of HCC cells and abrogated the potency of tumor formation in vivo. CONCLUSION: Targeting CALM2 may be a molecular strategy for both primary HCC treatment and prevention of metastasis or recurrence.


Assuntos
Calmodulina/fisiologia , Carcinoma Hepatocelular/patologia , Fator de Transcrição E2F5/fisiologia , Neoplasias Hepáticas/patologia , Apoptose/efeitos dos fármacos , Calmodulina/antagonistas & inibidores , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Metástase Neoplásica , Células-Tronco Neoplásicas/fisiologia
10.
Genet Test Mol Biomarkers ; 25(3): 179-186, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33734894

RESUMO

Background: E2F5 is a transcription factor that is overexpressed in the early stages of ovarian cancer and has been suggested as a potential biomarker for early detection. In this study, we aimed to examine the role of E2F5 in invasion and proliferation of ovarian cancer cells. Materials and Methods: We performed cell viability, colony formation, and invasion assays using ovarian cancer cells treated with siRNA to knock down the E2F5 gene. The regulatory effects of E2F5 on proteins involved in the apoptotic, Wnt, Hippo, and retinoblastoma signaling pathways were evaluated by western blotting following E2F5 repression. In addition, we analyzed data available on Gene Expression Profiling Interactive Analysis for correlations between E2F5 and YAP, ß-catenin, cyclin D1, cdk4, and caspase-9. Results: E2F5 was highly expressed in ovarian cancer cell lines and samples when compared to the nonmalignant tissues. Downregulation of E2F5 inhibited cell viability and invasion and promoted the phosphorylation of YAP, GSK-3-ß, ß-catenin, and retinoblastoma. However, cyclin D1, cdk4, and caspase-9 were downregulated when compared to control. Conclusion: Overall, E2F5 promotes ovarian carcinogenesis via the regulation of Hippo and Wnt pathways.


Assuntos
Fator de Transcrição E2F5/metabolismo , Neoplasias Ovarianas/metabolismo , Apoptose/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Ciclina D1/genética , Bases de Dados Genéticas , Fator de Transcrição E2F5/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Via de Sinalização Hippo , Humanos , Invasividade Neoplásica/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
11.
Anticancer Drugs ; 32(6): 602-613, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33595944

RESUMO

Long non-coding RNAs have the regulatory roles in different kinds of human cancers. The key point of this study was to research the functional mechanisms of urothelial carcinoma associated 1 (UCA1) in the development of osteosarcoma. Quantitative real-time PCR was adopted for the expression detection of UCA1, microRNA-513b-5p (miR-513b-5p) and E2F transcription factor 5 (E2F5). The target relation was verified via dual-luciferase reporter assay and RNA pull-down assay. Cell proliferation was evaluated using Cell Counting Kit-8 and colony formation assays. Transwell assay was applied to assess cell migration and invasion. Western blot was performed for protein examination. Xenograft experiment was used to explore the effect of UCA1 on osteosarcoma in vivo. UCA1 expression was enhanced while miR-513b-5p was refrained in osteosarcoma tissues and cells. MiR-513b-5p was a target of UCA1. Inhibition of UCA1 or overexpression of miR-513b-5p suppressed osteosarcoma cell proliferation, migration and invasion. E2F5 was identified as a downstream gene of miR-513b-5p. MiR-513b-5p inhibitor or E2F5 overexpression rescued the progression inhibition of osteosarcoma by UCA1 knockdown, and UCA1 regulated E2F5 and Cyclin E expression by targeting miR-513b-5p. Downregulation of UCA1 restrained the tumorigenesis of osteosarcoma in vivo through the miR-513b-5p/E2F5 axis. Collectively, knockdown of UCA1 inhibited tumorigenesis and metastasis of osteosarcoma via regulating the miR-513b-5p/E2F5 axis. UCA1 might be a biological indicator in the progression and treatment of osteosarcoma.


Assuntos
Neoplasias Ósseas/metabolismo , Fator de Transcrição E2F5/metabolismo , MicroRNAs/metabolismo , Osteossarcoma/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Carcinogênese , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Progressão da Doença , Regulação para Baixo , Fator de Transcrição E2F5/genética , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica , Metástase Neoplásica , Osteossarcoma/genética , Osteossarcoma/patologia , RNA Longo não Codificante/genética , Transdução de Sinais
12.
J Exp Clin Cancer Res ; 40(1): 2, 2021 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-33390186

RESUMO

BACKGROUND: Both E2F transcription factor and cyclin-dependent kinases (CDKs), which increase or decrease E2F activity by phosphorylating E2F or its partner, are involved in the control of cell proliferation, and some circRNAs and miRNAs regulate the expression of E2F and CDKs. However, little is known about whether dysregulation among E2Fs, CDKs, circRNAs and miRNAs occurs in human PCa. METHODS: The expression levels of CDK13 in PCa tissues and different cell lines were determined by quantitative real-time PCR and Western blot analysis. In vitro and in vivo assays were preformed to explore the biological effects of CDK13 in PCa cells. Co-immunoprecipitation anlysis coupled with mass spectrometry was used to identify E2F5 interaction with CDK13. A CRISPR-Cas9 complex was used to activate endogenous CDK13 and circCDK13 expression. Furthermore, the mechanism of circCDK13 was investigated by using loss-of-function and gain-of-function assays in vitro and in vivo. RESULTS: Here we show that CDK13 is significantly upregulated in human PCa tissues. CDK13 depletion and overexpression in PCa cells decrease and increase, respectively, cell proliferation, and the pro-proliferation effect of CDK13 is strengthened by its interaction with E2F5. Mechanistically, transcriptional activation of endogenous CDK13, but not the forced expression of CDK13 by its expression vector, remarkably promotes E2F5 protein expression by facilitating circCDK13 formation. Further, the upregulation of E2F5 enhances CDK13 transcription and promotes circCDK13 biogenesis, which in turn sponges miR-212-5p/449a and thus relieves their repression of the E2F5 expression, subsequently leading to the upregulation of E2F5 expression and PCa cell proliferation. CONCLUSIONS: These findings suggest that CDK13 upregulation-induced formation of the positive feedback loop among circCDK13, miR-212-5p/miR-449a and E2F5 is responsible for PCa development. Targeting this newly identified regulatory axis may provide therapeutic benefit against PCa progression and drug resistance.


Assuntos
Proteína Quinase CDC2/metabolismo , Fator de Transcrição E2F5/metabolismo , MicroRNAs/metabolismo , Neoplasias da Próstata/metabolismo , Proteína Quinase CDC2/genética , Proliferação de Células/fisiologia , Fator de Transcrição E2F5/genética , Retroalimentação , Feminino , Humanos , Masculino , MicroRNAs/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transfecção , Regulação para Cima
13.
Panminerva Med ; 63(3): 336-342, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32414231

RESUMO

BACKGROUND: MicroRNA-1271-5p (miR-1271-5p) has been reported to participate in the progression of many human cancers. However, the role of miR-1271-5p still remains unclear in ovarian cancer (OC). Therefore, we explored the effect of miR-1271-5p on the development of OC in present study. METHODS: We measured the miR-1271-5p expression via the qRT-PCR assay. Then the function of miR-1271-5p was analyzed through MTT and Transwell assays. The relationship among miR-1271-5p and E2F5 was verified by dual luciferase assay. The protein expression levels were examined through western blot. RESULTS: MiR-1271-5p was downregulated in OC tissues which predicted poor prognosis of OC patients. Moreover, E2F5 was a direct target of miR-1271-5p in OC. And miR-1271-5p suppressed cell proliferation, migration and invasion in OC through targeting E2F5. Furthermore, E2F5 was upregulated in OC tissues which predicted poor prognosis of OC patients. Besides that, miR-1271-5p suppressed EMT and mTOR pathway in OC. CONCLUSIONS: MiR-1271-5p inhibited the tumorigenesis of OC through targeting E2F5 and negatively regulated the mTOR signaling pathway.


Assuntos
MicroRNAs , Neoplasias Ovarianas , Carcinogênese , Fator de Transcrição E2F5 , Feminino , Humanos , MicroRNAs/genética , Neoplasias Ovarianas/genética , Transdução de Sinais , Serina-Treonina Quinases TOR
14.
Oncol Rep ; 44(5): 2241-2252, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33000282

RESUMO

E2F transcription factor 5 (E2F5) is a member of the E2F family of transcription factors, which are involved in regulation of various cellular processes, including cellular proliferation, apoptosis, differentiation and DNA damage response. Previously, we reported that E2F5 was aberrantly overexpressed in estrogen receptor (ER)­negative breast cancer, especially in triple­negative breast cancer (TNBC). In the present study, it was revealed that E2F5 gene silencing caused a significant reduction in the proliferation rate of breast cancer MCF7 (ER­positive luminal­type) and MDA­MB­231 (TNBC­type) cells. Additional experiments demonstrated that E2F5 knockdown triggered cell death of MCF7 cells but not MDA­MB­231 cells. As MCF7 and MDA­MB­231 cells carry wild­type and mutant TP53, respectively, and BT474 (ER­negative, HER2­positive type) carrying mutant TP53 exhibited similar results to MDA­MB­231, the possible effects of E2F5 gene depletion on cell death­related TP53­target gene expression were examined. Real­time RT­qPCR analysis revealed that knockdown of E2F5 in MCF7 cells stimulated cell death­related transcription of TP53­target genes such as BAX, NOXA and PUMA. For MDA­MB­231 and BT474 cells, E2F5 gene silencing revealed marginal effects on the expression of TP53 target genes. In addition, silencing of TP53 abrogated the effect of E2F5 silencing in MCF7 cells. Collectively, the present results indicated that E2F5 participated in the carcinogenesis of breast cancer carrying wild­type TP53 through suppression of TP53, while E2F5 had a pro­proliferative but not anti­apoptotic effect on breast cancer with TP53 mutation.


Assuntos
Carcinogênese/genética , Fator de Transcrição E2F5/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias de Mama Triplo Negativas/genética , Proteína Supressora de Tumor p53/metabolismo , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Fator de Transcrição E2F5/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/genética
15.
BMC Cancer ; 20(1): 374, 2020 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-32366234

RESUMO

BACKGROUND: The adhesion molecule, FAT4, has a tumor suppressor function with a critical role in the epithelial-to-mesenchymal-transition (EMT) and anti-malignant growth in several cancers. No study has investigated yet its role in epithelial ovarian cancer (EOC) progression. In the present study, we examined the role of FAT4 in proliferation and metastasis, and its mechanisms of interaction in these processes. METHODS: We have performed cell viability, colony formation, and invasion assays in ovarian cancer cells treated with siRNA to knockdown FAT4 gene expression. The regulatory effects of FAT4 on proteins involved in apoptotic, Wnt, Hippo, and retinoblastoma signaling pathways were evaluated by Western blotting following FAT4 repression. Also, 426 ovarian tumor samples and 88 non-tumor samples from the Gene Expression Profiling Interactive Analysis (GEPIA) database were analyzed for the expression of FAT4. Pearson's correlation was performed to determine the correlation between FAT4 and the E2F5, cyclin D1, cdk4, and caspase 9 expressions. RESULTS: Lower expression of FAT4 was observed in ovarian cancer cell lines and human samples as compared to non-malignant tissues. This down-regulation seems to enhance cell viability, invasion, and colony formation. Silencing FAT4 resulted in the upregulation of E2F5, vimentin, YAP, ß-catenin, cyclin D1, cdk4, and Bcl2, and in the downregulation of GSK-3-ß, and caspase 9 when compared to control. Furthermore, regulatory effects of FAT4 on the EMT and aggressive phenotype seem to occur through Hippo, Wnt, and cell cycle pathways. CONCLUSION: FAT4 downregulation promotes increased growth and invasion through the activation of Hippo and Wnt-ß-catenin pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caderinas/antagonistas & inibidores , Fator de Transcrição E2F5/metabolismo , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/patologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/antagonistas & inibidores , beta Catenina/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Genes Supressores de Tumor , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Sinalização YAP
16.
Carcinogenesis ; 41(12): 1767-1780, 2020 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-32386317

RESUMO

Previously, our laboratory demonstrated that a deregulated E2F5/p38/SMAD3 axis was associated with uncontrolled cellular proliferation in prostate cancer (PCa). Here, we investigate the role of E2F5 in PCa in further details. RNAi-mediated E2F5 knockdown and pathway-focused gene expression profiling in PC3 cells identified TFPI2 as a downstream target of E2F5. Manipulation of E2F5 expression was also found to alter MMP-2 and MMP-9 levels as detected by Proteome Profiler array, western blot and reverse transcription coupled quantitative polymerase chain reaction Site-directed mutagenesis, dual-luciferase assays and chromatin immunoprecipitation with anti-E2F5-IgG coupled with qPCR confirmed recruitment of E2F5 on TFPI2, MMP-2 and MMP-9 promoters. RNAi-mediated knockdown of E2F5 expression in PC3 caused a significant alteration of cell migration while that of TFFI2 resulted in a modest change. Abrogation of E2F5 and TFPI2 expression was associated with significant changes in the gelatinolytic activity of active forms of MMP-2 and MMP-9. Moreover, E2F5, MMP-2 and MMP-9 levels were elevated in biopsies of PCa patients relative to that of benign hyperplasia, while TFPI2 expression was reduced. MMP-9 was coimmunoprecipitated with anti-TFPI2-IgG in PCa tissue samples suggesting a direct interaction between the proteins. Finally, artemisinin treatment in PC3 cells repressed E2F5 along with MMP-2/MMP-9 while triggering TFPI2 expression which alleviated PC3 aggressiveness possibly through inhibition of MMP activities. Together, our study reinstates an oncogenic role of E2F5 which operates as a dual-function transcription factor for its targets TFPI2, MMP-2 and MMP-9 and promotes cellular invasiveness. This study also indicates a therapeutic potential of artemisinin, a natural compound which acts by correcting dysfunctional E2F5/TFPI2/MMP axis in PCa.


Assuntos
Biomarcadores Tumorais/metabolismo , Fator de Transcrição E2F5/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias da Próstata/patologia , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Fator de Transcrição E2F5/genética , Glicoproteínas/genética , Humanos , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Invasividade Neoplásica , Prognóstico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Células Tumorais Cultivadas
17.
Cell Cycle ; 19(13): 1611-1620, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32420810

RESUMO

BACKGROUND/AIMS: CircABCB10 function as an endogenous miRNA sponge plays an important role in various tumors. This experimental design was based on circABCB10 to explore the pathogenesis of non-small cell lung cancer (NSCLC). Methods: CircRNA microarray was used to examine circRNA expression profiles in lung cancer from 3 NSCLC patients and paired healthy lung tissues. The expression of circABCB10 and miR-584-5p was detected by q-PCR. CCK-8, colony formation, and transwell assays to study the circABCB10 effects on tumor cell growth and cell migration invasiveness. To validate downstream target genes of circABCB10 and miR-584-5p detected by luciferase reporter assays. RT-qPCR and Western blotting were used to study E2F5 expression. The tumor growth was detected by nude mice in vivo. Results: We analyzed the human circRNA expression profile in NSCLC tissues. CircABCB10 was identified as a circRNA that increased in NSCLC tissues. CircABCB10 was noticeably raised in NSCLC, and high circABCB10 expression was related to low survival in NSCLC patients. Silencing of circABCB10 suppressed non-small cell lung cancer cell migration, cell proliferation, and invasion.CircABCB10 can act as a sponge of miR-584-5p to up-regulate E2F5 expression level. E2F5 knockdown or overexpress of miR-584-5p gene reversed the circABCB10 who has carcinogenic effects. There was a negative correlation expression between the circABCB10 and miR-584-5p gene, and There was a positive relationship between the expression of circABCB10 and E2F5 in NSCLC tumors. Conclusion: CircABCB10 promoted the progression of NSCLC by modulating the miR-584-5p/E2F5 axis. ABBREVIATION: NSCLC: non-small cell lung cancer; circ RNA: circular RNA; miRNA: micro RNA.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Progressão da Doença , Fator de Transcrição E2F5/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , MicroRNAs/metabolismo , RNA Circular/metabolismo , Idoso , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Fator de Transcrição E2F5/metabolismo , Feminino , Inativação Gênica , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Prognóstico , RNA Circular/genética
18.
PLoS Genet ; 16(3): e1008655, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32196499

RESUMO

E2f5 is a member of the E2f family of transcription factors that play essential roles during many cellular processes. E2f5 was initially characterized as a transcriptional repressor in cell proliferation studies through its interaction with the Retinoblastoma (Rb) protein for inhibition of target gene transcription. However, the precise roles of E2f5 during embryonic and post-embryonic development remain incompletely investigated. Here, we report that zebrafish E2f5 plays critical roles during spermatogenesis and multiciliated cell (MCC) differentiation. Zebrafish e2f5 mutants develop exclusively as infertile males. In the mutants, spermatogenesis is arrested at the zygotene stage due to homologous recombination (HR) defects, which finally leads to germ cell apoptosis. Inhibition of cell apoptosis in e2f5;tp53 double mutants rescued ovarian development, although oocytes generated from the double mutants were still abnormal, characterized by aberrant distribution of nucleoli. Using transcriptome analysis, we identified dmc1, which encodes an essential meiotic recombination protein, as the major target gene of E2f5 during spermatogenesis. E2f5 can bind to the promoter of dmc1 to promote HR, and overexpression of dmc1 significantly increased the fertilization rate of e2f5 mutant males. Besides gametogenesis defects, e2f5 mutants failed to develop MCCs in the nose and pronephric ducts during early embryonic stages, but these cells recovered later due to redundancy with E2f4. Moreover, we demonstrate that ion transporting principal cells in the pronephric ducts, which remain intercalated with the MCCs, do not contain motile cilia in wild-type embryos, while they generate single motile cilia in the absence of E2f5 activity. In line with this, we further show that E2f5 activates the Notch pathway gene jagged2b (jag2b) to inhibit the acquisition of MCC fate as well as motile cilia differentiation by the neighboring principal cells. Taken together, our data suggest that E2f5 can function as a versatile transcriptional activator and identify novel roles of the protein in spermatogenesis as well as MCC differentiation during zebrafish development.


Assuntos
Fator de Transcrição E2F5/metabolismo , Espermatogênese/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteínas de Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Cílios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição E2F5/genética , Masculino , Receptores Notch/metabolismo , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
19.
Mol Med Rep ; 20(2): 2012-2020, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31257477

RESUMO

The dysregulated behavior of vascular smooth muscle cells (VSMCs) serves an important role in the pathogenesis of cardiovascular diseases in diabetes. The present study aimed to investigate the effects of microRNA (miR)­132 on the proliferation and migration of VSMCs under high glucose conditions to mimic diabetes. We observed that the expression of miR­132 was significantly decreased and that of E2F transcription factor 5 (E2F5) was upregulated in high glucose (HG)­treated VSMCs or those obtained from diabetic rats. A dual luciferase reporter gene assay revealed that miR­132 could specifically bind to the 3'­untranslated region of E2F5 and significantly suppress the luciferase activity. The proliferation and migration of diabetic rat or HG­treated VSMCs were increased compared with non­diabetic rat VSMCs and those under normal glucose conditions. Upregulation of miR­132 significantly inhibited the proliferation and migration of diabetic rat VSMCs; similar effects were observed following E2F5 downregulation. The inhibitory effects of miR­132 on the proliferation and migration of HG­treated VSMCs could be reversed by E2F5 overexpression. In conclusion, miR­132 was proposed to inhibit the proliferation and migration of diabetic rat or high­glucose­treated VSMCs by targeting E2F5. The findings of the present study suggested that increasing the expression of miR­132 may serve as a novel therapeutic approach to inhibit the progression of cardiovascular disease in diabetes.


Assuntos
Fator de Transcrição E2F5/genética , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Regulação da Expressão Gênica/genética , Glucose/farmacologia , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/genética , Ratos
20.
J Cell Physiol ; 234(11): 19523-19538, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30997692

RESUMO

Periodontal ligament stem cells (PDLSCs) are characterized by multiple differentiation potential and potent self-renewal ability, yet much remains to be elucidated that what determines these properties. Long noncoding RNAs (lncRNAs) have been suggested to involve in multiple biological process under physiological and pathological conditions, including osteogenic differentiation. In the present study, we performed comprehensive lncRNA profiling by lncRNA microarray analysis and identified prostate cancer-associated ncRNA transcript-1 (lncPCAT1) was gradually increased in PDLSCs during consecutive osteogenic induction, and it could further positively regulate the osteogenic differentiation both in vitro and in vivo, whereas lncPCAT1 inhibition led to suppressed osteogenic differentiation. Thereafter, we inferred a predicted interaction between lncPCAT1 and miR-106a-5p and then confirmed the direct binding sites of miR-106a-5p on lncPCAT1. Although miR-106a-5p upregulation led to decreased osteogenic differentiation, lncPCAT1 overexpression could reverse its suppression, indicating that lncPCAT1 act as a competing endogenous RNA for miR-106a-5p. Moreover, lncPCAT1 could sponge miR-106a-5p to upregulate miR-106a-5p-targeted gene BMP2, which was a crucial gene involved in osteogenic differentiation. Interestingly, we found that E2F5, another target of miR-106a-5p, could bind to the promoter of lncPCAT1 and then form a feed-forward regulatory network targeting BMP2. In conclusion, our study provided a novel lncRNA-miRNA feed-forward regulatory network and a promising target to modulate the osteogenic differentiation of PDLSCs.


Assuntos
Fator de Transcrição E2F5/genética , MicroRNAs/genética , Osteogênese/genética , Ligamento Periodontal/crescimento & desenvolvimento , RNA Longo não Codificante/genética , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/genética , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Ligamento Periodontal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA