Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Int J Mol Sci ; 18(3)2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28257042

RESUMO

We previously identified proline-rich protein 11 (PRR11) as a novel cancer-related gene that is implicated in the regulation of cell cycle and tumorigenesis. Our recent study demonstrated that PRR11 and its adjacent gene, kinetochore associated 2 (SKA2), constitute a classic head-to-head gene pair that is coordinately regulated by nuclear factor Y (NF-Y). In the present study, we further show that the PRR11-SKA2 bidirectional transcription unit is an indirect target of the tumor suppressor p53. A luciferase reporter assay revealed that overexpression of wild type p53, but not mutant p53, significantly represses the basal activity and NF-Y mediated transactivation of the PRR11-SKA2 bidirectional promoter. Deletion and mutation analysis of the PRR11-SKA2 promoter revealed that p53-mediated PRR11-SKA2 repression is dependent on the presence of functional NF-Y binding sites. Furthermore, a co-immunoprecipitation assay revealed that p53 associates with NF-Y in lung cancer cells, and a chromatin immunoprecipitation assay showed that p53 represses PRR11-SKA2 transcription by reducing the binding amount of NF-Y in the PRR11-SKA2 promoter region. Consistently, the ability of p53 to downregulate PRR11-SKA2 transcription was significantly attenuated upon siRNA-mediated depletion of nuclear factor Y subunit beta (NF-YB). Notably, lung cancer patients with lower expression of either PRR11 or SKA2 along with wild type p53 exhibited the best overall survival compared with others with p53 mutation and/or higher expression of either PRR11 or SKA2. Taken together, our results demonstrate that p53 negatively regulates the expression of the PRR11-SKA2 bidirectional transcription unit through NF-Y, suggesting that the inability to repress the PRR11-SKA2 bidirectional transcription unit after loss of p53 might contribute to tumorigenesis.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Proteínas Cromossômicas não Histona/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/mortalidade , Prognóstico , Regiões Promotoras Genéticas , Ligação Proteica
2.
Biochim Biophys Acta ; 1849(9): 1133-44, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26162986

RESUMO

Head-to-head gene pairs represent a unique feature of gene organization in eukaryotes, accounting for >10% of genes in the human genome. Identification and functional analysis of such gene pairs is only in its infancy. Recently, we identified PRR11 as a novel cancer-related gene that is implicated in cell cycle and lung cancer. Here we demonstrate that PRR11 is oriented in a head-to-head configuration with its neighboring gene, SKA2. 5'-RACE assay revealed that the intergenic spacer region between the two genes is <500 bp. Serial luciferase reporter assays demonstrated that a minimal 80-bp intergenic region functions as a core bidirectional promoter to drive basal transcription in both the PRR11 and SKA2 orientations. EMSA and ChIP assays demonstrated that NF-Y binds to and directly transactivates the PRR11-SKA2 bidirectional promoter. SiRNA-mediated NF-Y depletion significantly downregulated PRR11 and SKA2 expression. Expression of both PRR11 and SKA2 was significantly upregulated in lung cancer. Expression of the two genes was highly correlated with each other and with NF-Y expression. Remarkably, high expression of both PRR11 and SKA2 was associated with poorer prognosis in lung cancer patients compared with high expression of one gene or low expression of both genes. Knockdown of PRR11 and/or SKA2 remarkably reduced cell proliferation, migration, and invasion in lung cancer cells. Thus, the PRR11-SKA2 bidirectional transcription unit, which is a novel direct target of NF-Y, is essential for the accelerated proliferation and motility of lung cancer cells and may represent a potential target in the diagnosis and/or treatment of human lung cancer.


Assuntos
Fator de Ligação a CCAAT/fisiologia , Proteínas Cromossômicas não Histona/genética , Regulação da Expressão Gênica , Neoplasias Pulmonares/genética , Regiões Promotoras Genéticas , Proteínas/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Cromatina/genética , DNA/genética , Humanos , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica
3.
Saudi Pharm J ; 24(3): 279-85, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27275115

RESUMO

This study focuses on investigation of cryptorchidism induced by flutamide (Flu) and its histopathological damage, and detects retinoic acid concentration in testicle tissue, in order to find a new method for clinical treatment to infertility caused by cryptorchidism. Twenty SD (Sprague Dawley) pregnant rats were randomly divided into Flu cryptorchidism group (n = 10) and normal control group (n = 10). HE stained for observing morphological difference. Transmission electron microscope (TEM) was used for observing the tight junction structure between Sertoli cells. Epididymal caudal sperms were counted and observed in morphology. The expression of stimulated by retinoic acid gene 8 (Stra8) was detected using immunohistochemistry, western blot, and Q-PCR. High performance liquid chromatography (HPLC) analysis was made on retinoic acid content. Sperm count and morphology observation confirmed cryptorchidism group was lower than normal group in sperm quantity and quality. The observation by TEM showed a loose structure of tight junctions between Sertoli cells. Immunohistochemistry, western blot, and Q-PCR showed that cryptorchidism group was significantly lower than normal group in the expression of Stra8. HPLC showed that retinoic acid content was significantly lower in cryptorchid testis than in normal testis. In the cryptorchidism model, retinoic acid content in testicular tissue has a significant reduction; testicles have significant pathological changes; damage exists in the structure of tight junctions between Sertoli cells; Stra8 expression has a significant reduction, perhaps mainly contributing to spermatogenesis disorder.

4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(8): 1036-40, 2016 Aug.
Artigo em Zh | MEDLINE | ID: mdl-27412932

RESUMO

Objective To investigate the effect of small interfering RNA (siRNA)-mediated silencing of the Bmi-1 gene on cell proliferation and invasion of MCF-7 human mammary carcinoma cell line and the potential molecular mechanisms. Methods Real-time quantitative PCR was used to detect the levels of Bmi-1 mRNA in the paired breast cancer and adjacent noncancerous breast tissues which were confirmed by pathological diagnosis. Bmi-1-siRNA was transfected into MCF-7 cells by a Lipofectamine(R) RNAiMAX transfection reagent. Flow cytometry was used to detect cell cycle and apoptosis of MCF-7 cells transfected by Bmi-1-siRNA. Western blotting was performed to detect the protein levels of P21, Bax and Bcl-2. Matrigel Transwell(TM) invasion assay was used to determine the cell invasion of MCF-7 cells with Bmi-1 silencing. The protein levels of E-cadherin, N-cadherin, vimentin were tested by Western blotting. Results The expression of Bmi-1 mRNA in the breast cancer tissues was higher than that in the adjacent noncancerous breast tissues. Bmi-1 silencing significantly suppressed the cell growth, arrested the cells in the G1/S phase and promoted the apoptosis of MCF-7 cells. Compared with blank control group or negative control group, the Bmi-1-silenced group showed the increased expressions of P21 and Bax and the decreased expression of Bcl-2. In addition, Bmi-1 silencing significantly suppressed the cell invasion and promoted the expression of E-cadherin as well as downregulated the expressions of N-cadherin and vimentin in MCF-7 cells. Conclusion The invasion of MCF-7 cells can be inhibited by Bmi-1 silencing, of which the molecular regulation mechanism might be associated with the inhibition of tumor cell epithelial-mesenchymal transition.


Assuntos
Apoptose/genética , Movimento Celular/genética , Complexo Repressor Polycomb 1/genética , Interferência de RNA , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citometria de Fluxo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Invasividade Neoplásica , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vimentina/metabolismo , Proteína X Associada a bcl-2/metabolismo
5.
Int J Biochem Cell Biol ; 45(3): 645-56, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23246489

RESUMO

Identification and functional analysis of novel potential cancer-associated genes is of great importance for developing diagnostic, preventive and therapeutic strategies for cancer treatment and management. In the present study, we isolated and identified a novel gene, proline-rich protein 11 (PRR11), implicated in both cell cycle progression and lung cancer. Our results showed that PRR11 was periodically expressed in a cell cycle-dependent manner, and RNAi-mediated silencing of PRR11 caused significant S phase arrest as well as growth retardation in HeLa cells. Moreover, PRR11 was overexpressed at both mRNA and protein levels in lung cancer tissues as compared with normal lung tissues. Large scale in silico analysis of clinical microarray datasets also indicated that high expression of PRR11 was significantly associated with poor prognosis in lung cancer patients. RNAi-mediated silencing of PRR11 caused S phase arrest, suppressed cellular proliferation, colony formation ability in lung cancer cells and inhibited tumorigenic potential in nude mice. Knockdown of PRR11 also inhibited cell migration and invasion ability in lung cancer cells. Furthermore, microarray analysis revealed that PRR11 knockdown caused the dysregulation of multiple critical pathways and various important genes involved in cell cycle, tumorigenesis and metastasis (e.g. CCNA1, RRM1, MAP4K4 and EPB41L3). Taken together, our results strongly demonstrated that this newly identified gene, PRR11, had a critical role in both cell cycle progression and tumorigenesis, and might serve as a novel potential target in the diagnosis and/or treatment of human lung cancer.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Transformação Celular Neoplásica/genética , Neoplasias Pulmonares/genética , Domínios Proteicos Ricos em Prolina/genética , Proteínas/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA