Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 41(6): 3367-3376, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31002360

RESUMO

Aberrantly expressed miRNAs play a crucial role in the progression of lung adenocarcinoma. However, to date, the role of miR­888 in lung adenocarcinoma progression is unclear. In the present study, the biological function of miR­888 and its underlying mechanism in lung adenocarcinoma progression were explored. RT­qPCR was performed to detect the expression of miR­888 in 38 matched lung adenocarcinoma samples respectively. Next, the effects of miR­888 on the proliferation, invasion and migration of lung adenocarcinoma A549 cells were evaluated by a series of gain­ and loss­of­function assays. Our results revealed that miR­888 was significantly upregulated in lung adenocarcinoma tissues, and its expression was markedly associated with clinical staging in patients. Moreover, ectopic expression of miR­888 in vitro was revealed to function as a double­edged sword in the progression of lung adenocarcinoma A549 cells by targeting multiple targets. Overexpression of miR­888 promoted the invasion and migration of lung adenocarcinoma A549 cells by targeting E­cadherin and tissue inhibitor of metalloproteinase 2. In addition, ectopic expression of miR­888 inhibited the proliferation of lung adenocarcinoma A549 cells by targeting cell division cycle 7 (CDC7). In addition, the immunohistochemical results and The Cancer Genome Atlas (TCGA) database revealed that CDC7 was significantly upregulated in lung adenocarcinoma tissues, suggesting that miR­888 may function as an oncogene in the progression of lung adenocarcinoma patients, and the miR­888/CDC7 axis was not the dominant pathway for CDC7 regulation in patients with lung adenocarcinoma. In conclusion, our findings indicated that miR­888 may act as a potential new therapeutic target for patients with lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/genética , Biomarcadores Tumorais/genética , Proteínas de Ciclo Celular/genética , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , Células A549 , Adenocarcinoma de Pulmão/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Caderinas/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Pessoa de Meia-Idade , Inibidor Tecidual de Metaloproteinase-2/genética
2.
Onco Targets Ther ; 12: 63-74, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30588031

RESUMO

PURPOSE: The cell division cycle 7 (CDC7) is a serine/threonine kinase that is essential for DNA replication in human cells which has been identified to play a critical role in multiple cancer types. However, the expression and clinical significance of CDC7 in ESCC has never been reported. PATIENTS AND METHODS: CDC7 expression was detected in 30 ESCC and matched adjacent normal tissues, and a series of loss-of-function and gain-of-function assays were performed to evaluate the effects of CDC7 on the proliferation, migration and invasion, and chemoresistance of ESCC cells. RESULTS: The results showed that CDC7 was highly expressed in ESCC tissues compared with matched adjacent normal tissues. Functional studies demonstrated that knockdown of CDC7 inhibited proliferation by arresting ESCC cells in the G0/G1 phase and inducing apoptosis. Knockdown of CDC7 also inhibited cell migration and invasion in ESCC cells. Furthermore, knockdown of CDC7 sensitized ESCC cells to Cis and 5-FU. CONCLUSION: Our results suggest that CDC7 is highly expressed in ESCC tissues, and silencing CDC7 enhances chemosensitivity of ESCC cells, providing a new avenue for ESCC therapy.

3.
Biochem Biophys Res Commun ; 478(2): 676-82, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27498003

RESUMO

MicroRNAs (miRNAs) are potent post-transcriptional regulators of gene expression and play roles in DNA damage response (DDR). PLK1 is identified as a modulator of DNA damage checkpoint. Although down-regulation of PLK1 by certain microRNAs has been reported, little is known about the interplay between PLK1 and miR-509-3-5p in DDR. Here we have demonstrated that miR-509-3-5p repressed PLK1 expression by targeting PLK1 3'-UTR, thereby causing mitotic aberration and growth arrest of human lung cancer A549 cells. Repression of PLK1 by miR-509-3-5p was further evidenced by over-expression of miR-509-3-5p in A549, HepG2 and HCT116p53(-/-) cancer cells, in which PLK1 protein was suppressed. Consistently, miR-509-3-5p was stimulated, while PLK1 protein was down-regulated in A549 cells exposed to CIS and ADR, suggesting that suppression of PLK1 by miR-509-3-5p is a component of CIS/ADR-induced DDR pathway. Flow cytometry and immunofluorescence labeling showed that over-expression of miR-509-3-5p in A549 induced G2/M arrest and aberrant mitosis characterized by abnormal bipolar mitotic spindles, condensed chromosomes, lagging DNA and chromosome bridges. In addition, over-expression of miR-509-3-5p markedly blocked A549 cell proliferation and sensitized the cells to CIS and ADR treatment. Taken together, miR-509-3-5p is a feasible suppressor for cancer by targeting PLK1. Our data may provide aid in potential design of combined chemotherapy and in our better understanding of the roles of microRNAs in response to DNA damage.


Assuntos
Proteínas de Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Regiões 3' não Traduzidas , Células A549 , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Sequência de Bases , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células HCT116 , Células Hep G2 , Humanos , MicroRNAs/metabolismo , Mitose/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Quinase 1 Polo-Like
4.
Mol Cell Biochem ; 399(1-2): 179-88, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312903

RESUMO

The p53R2 gene encoding a small subunit of the ribonucleotide reductase has been identified as a p53-inducible gene. Although this gene is discovered as a target for p53 family proteins, the mechanism underlying p53R2 induction by DNA damage in p53-defiencient cells remains to be elucidated. In this study, we demonstrate that transcription factor E2F1 regulates the p53R2 gene expression in p53-deficient cells. We found that p53R2 was a target for E2F1 in DNA damage response (DDR), because ectopic expression of E2F1 in HCT116-p53(-/-) cells resulted in the increase of p53R2 mRNA and protein expression, and silencing E2F1 diminished its basic expression. Combination of luciferase reporter assay with overexpression or knockdown of E2F1 revealed that E2F1 directly activates the p53R2 gene. Chromatin immunoprecipitation (ChIP) assay showed E2F1 directly bound to the site (TTTGGCGG) at position -684 to -677 of the promoter under E2F1 overexpression or adriamycin (ADR) exposure. Moreover, silencing p53R2 could enhance apoptotic cell death in both HCT116-p53(-/-) and HCT116-p53(+/+) compared to ADR exposure, indicating that p53R2 may protect cancer cell from ADR-induced apoptosis. Together, we have identified a new role of E2F1 in the regulation of p53R2 expression in DDR, and silencing p53R2 may sensitize cancer cells to ADR-induced apoptosis. Our data support the notion that p53R2 is a potential target for cancer therapy. The involvement of E2F1-dependent p53R2 activation in DDR will provide further insight into the induction of p53R2 in p53-deficient cells. These data also give us a deeper understanding of E2F1 role in DDR.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fator de Transcrição E2F1/fisiologia , Ribonucleotídeo Redutases/metabolismo , Ativação Transcricional , Antibióticos Antineoplásicos/farmacologia , Apoptose , Sequência de Bases , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Dano ao DNA , Reparo do DNA , Doxorrubicina/farmacologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Ligação Proteica , Ribonucleotídeo Redutases/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
5.
Biochem Biophys Res Commun ; 450(1): 470-5, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24909689

RESUMO

DNA damage may regulate microRNA (miRNA) biosynthesis at the levels of miRNA transcription, processing and maturation. Although involvement of E2F1 in the regulation of miRNA gene activation in response to DNA damage has been documented, little is known about the role of E2F1 in miRNA processing. In this study we demonstrate that E2F1 enhances miR-630 biosynthesis under cisplatin (CIS) exposure through promoting DROSHA-mediated pri-miR-630 processing. Northern blot and RT-qPCR revealed that CIS exposure caused not only an increase in pri-miR-630 but also much more increase in pre-miR-630 and mature miR-630. The increases in pri-miR-630 and pre-miR-630 expression in unmatched proportion indicated that primary transcript processing was involved in CIS-stimulated miR-630 biosynthesis. Furthermore, combination of reporter enzyme assay with mutation and over-expression of E2F1 showed that induction of DROSHA promoted miR-630 expression, in which CIS-induced E2F1 activated DROSHA gene expression by recognizing and binding two E2F1 sites at the positions -214/-207 and -167/-160 of the DROSHA promoter. The increased binding of E2F1 to the DROSHA promoter in CIS-exposed cells was further evidenced by chromatin immunoprecipitation assay. Together, E2F1-regulated DROSHA promotes pri-miR-630 processing, thereby, contributes to CIS-stimulated miR-630 expression. The involvement of E2F1-dependent DROSHA activation in pri-miRNA processing under DNA damage stress will provide further insight into the regulation of miRNA biosynthesis. These data also give us a deeper understanding of E2F1 role in response to DNA damage.


Assuntos
Cisplatino/farmacologia , Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , MicroRNAs/biossíntese , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Ribonuclease III/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Humanos
6.
Exp Cell Res ; 319(20): 3104-15, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24076372

RESUMO

E2F1 is implicated in transcriptional activation of polo-like kinase-1 (PLK1), but yet the mechanism is not fully understood. PLK1 suppression plays an important checkpoint role in response to DNA damage. Suppression of the PLK1 gene by binding of p53 to upstream p53RE2 element in the promoter has been recently revealed. Here we report another mechanism, in which p53 interacts with E2F1 to form p53-E2F1-DNA complex repressing E2F1-dependent PLK1 expression. PLK1 was downregulated in cisplatin exposed HCT116p53(+/+) but not HCT116p53(-/-) cells, indicating p53-suppressed PLK1 upon DNA damage. Co-transfection and reporter enzyme assays revealed that p53 suppressed but E2F1 promoted PLK1 gene activation. 5'-Deletion and substitution mutations showed multiple positive cis-elements residing in the PLK1 promoter, of which at least two E2F1 sites at positions -75/-68 and -40/-32 were required for the full activity of the promoter. Combination of 5'-deletion and substitution mutations with over-expression of p53 showed that suppression of the PLK1 gene by p53 was E2F1-dependent: mutation of the E2F1 site at position -75/-68 partially abrogated suppression activity of p53; mutation of E2F1 site at position -40/-32 released from p53 suppression of PLK1 gene completely. Co-immunoprecipitation and electrophoretic mobility shift assay showed that DNA damage promoted p53-E2F1 interaction, thereby creating a p53-E2F1 complex assembly on the PLK1 promoter in vitro. The in vivo formation of p53-E2F1-PLK1 promoter complex upon DNA damage was further evidenced by chromatin immunoprecipitation (ChIP) and re-ChIP. In addition, we showed that suppression of PLK1 by p53 promoted apoptosis. Our data suggest that p53 may interact with E2F1 to form p53-E2F1-DNA complex suppressing E2F1-dependent PLK1 expression. The model of p53 action on E2F1-activated PLK1 gene may explain at least partly how p53 as a suppressor regulates the downstream effects of E2F1 in cellular stresses including DNA damage stress.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , DNA/metabolismo , Fator de Transcrição E2F1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , DNA/genética , Fator de Transcrição E2F1/antagonistas & inibidores , Fator de Transcrição E2F1/genética , Perfilação da Expressão Gênica , Células HCT116 , Humanos , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Quinase 1 Polo-Like
7.
Mol Cell Biochem ; 384(1-2): 187-96, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24037421

RESUMO

Although E2F1-mediated DNA double-stranded breaks (DSBs) and tetraploid have been extensively studied, the role of E2F1 in mitotic catastrophe is still unknown. We have previously shown that 8-chloro-adenosine (8-Cl-Ado) induces DNA DSBs and aberrant mitosis in human lung cancer cells, followed by delayed apoptosis. Here, we demonstrate that E2F1-mediated DNA damage is implicated in 8-Cl-Ado-induced chromosome missegregation and apoptosis in lung cancer H1299 cells. We showed that E2F1 was accumulated upon 8-Cl-Ado-induced DNA DSBs. Induction of E2F1 by 8-Cl-Ado caused DNA damage in cycling cells including M cells. In contrast, silencing of E2F1 expression decreased 8-Cl-Ado-induced DNA DSBs, particularly eliminated E2F1-mediated mitotic DNA damage. Over-expression of E2F1 and/or 8-Cl-Ado exposure resulted in aberrant mitotic spindles and chromosome segregation errors. Furthermore, over-expression of E2F1 expression enhanced 8-Cl-Ado-induced apoptosis. Together, our data indicate that E2F1-mediated DNA damage, in particular mitotic DNA damage, is an important fraction of 8-Cl-Ado-induced DNA damage, which is implicated in 8-Cl-Ado-induced mitotic catastrophe and delayed apoptosis. Induction of E2F1 by 8-Cl-Ado may contribute at least partly to the drug-inhibited proliferation of cancer cells.


Assuntos
2-Cloroadenosina/análogos & derivados , Apoptose/efeitos dos fármacos , Segregação de Cromossomos/genética , Fator de Transcrição E2F1/metabolismo , Neoplasias Pulmonares/genética , 2-Cloroadenosina/farmacologia , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Aberrações Cromossômicas , Segregação de Cromossomos/efeitos dos fármacos , Cromossomos/genética , Quebras de DNA de Cadeia Dupla , Regulação para Baixo , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Mitose/efeitos dos fármacos , Mitose/genética , Interferência de RNA , RNA Interferente Pequeno , Tetraploidia , Proteína Supressora de Tumor p14ARF/metabolismo
8.
Biochemistry (Mosc) ; 77(3): 261-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22803943

RESUMO

The E2F1 transcription factor is a well known regulator of cell proliferation and apoptosis, but its role in response to DNA damage is less clear. 8-Chloro-adenosine (8-Cl-Ado), a nucleoside analog, can inhibit proliferation in a variety of human tumor cells. However, it is still elusive how the agent acts on tumors. Here we show that A549 and H1299 cells formed DNA double-strand breaks after 8-Cl-Ado exposure, accompanied by E2F1 upregulation at protein level. Overexpressed wild-type (E2F1-wt) colocalized with double-strand break marker γ-H2AX and promoted G2/M arrest in 8-Cl-Ado-exposed A549 and H1299, while expressed S31A mutant of E2F1 (E2F1-mu) significantly reduced ability to accumulate at sites of DNA damage and G2/M arrest, suggesting that E2F1 is required for activating G2/M checkpoint pathway upon DNA damage. Transfection of either E2F1-wt or E2F1-mu plasmid promoted apoptosis in 8-Cl-Ado-exposed cells, indicating that 8-Cl-Ado may induce apoptosis in E2F1-dependent and E2F1-independent ways. These findings demonstrate that E2F1 plays a crucial role in 8-Cl-Ado-induced G2/M arrest but is dispensable for 8-Cl-Ado-induced apoptosis. These data also suggest that the mechanism of 8-Cl-Ado action is complicated.


Assuntos
2-Cloroadenosina/análogos & derivados , Adenocarcinoma/fisiopatologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fator de Transcrição E2F1/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Neoplasias Pulmonares/fisiopatologia , Pontos de Checagem da Fase M do Ciclo Celular , 2-Cloroadenosina/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma de Pulmão , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Fator de Transcrição E2F1/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...