Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clinics (Sao Paulo) ; 77: 100081, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36087568

RESUMO

OBJECTIVE: Long Non-Coding RNAs (LncRNAs) act as an indispensable role in cancer development. The study aimed to investigate the role and mechanism of lncRNA Small Nucleolar RNA Host Gene 1 (SNHG1) in Bladder Cancer (BC) progression. METHOD: The expression, prognostic value, diagnostic value, and correlation of SNHG1, Enhancer of Zeste 2 polycomb repressive complex 2 subunit (EZH2), and Kruppel Like Factor 2 (KLF2) were analyzed through bioinformatics analysis. The expression was also validated in BC tissues and cell lines. Besides, their regulation and binding were tested via qPCR, Western blot, Dual-Luciferase Reporter Assay (DLRA), Argonaute RISC catalytic component 2-RNA Immunoprecipitation (AGO2-RIP), and Chromatin Immunoprecipitation (ChIP). A xenograft model in nude mice was also established. RESULTS: SNHG1 was significantly overexpressed in BC tissues and cells. Importantly, SNHG1 was associated with poor survival, and ROC curves revealed high diagnostic values. Moreover, by CCK8, wound healing, transwell, and Western blot analysis, SNHG1 knockdown significantly inhibited the proliferation, migration, invasion, and epithelial-mesenchymal transition of BC cells. Additionally, in vivo experiments showed that silencing SNHG1 hindered tumorigenesis and tumor growth. Regarding mechanism, the results of AGO2-RIP, ChIP or DLRA showed that SNHG1 played different roles at diverse subcellular sites. In the cytoplasm, SNHG1 acted as a competing endogenous RNA for miR-137-3p to promote EZH2 expression. In the nucleus, SNHG1 could interact with EZH2 to inhibit KLF2 transcription. CONCLUSION: Our study elucidated that SNHG1 formed a regulatory network and played an oncogenic role in BC, which provided a novel therapeutic target for BC treatment.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , MicroRNAs , RNA Longo não Codificante , Neoplasias da Bexiga Urinária , Animais , Carcinogênese , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Nus , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias da Bexiga Urinária/genética
2.
Front Endocrinol (Lausanne) ; 13: 703733, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35197928

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive brain tumors. 17ß-estradiol (E2) increases proliferation, migration, and invasion of human GBM cells; however underlying mechanisms are no fully understood. Zeste 2 Enhancer Homologous enzyme (EZH2) is a methyltransferase part of Polycomb 2 repressor complex (PRC2). In GBM, EZH2 is overexpressed and involved in the cell cycle, migration, and invasion processes. We studied the role of EZH2 in the pro-oncogenic actions of E2 in human GBM cells. EZH2 gene silencing and pharmacological inhibition of EZH2 blocked proliferation, migration, and invasion of GBM cells induced by E2. We identified in silico additional putative estrogen response elements (EREs) at the EZH2 promoter, but E2 did not modify EZH2 expression. In silico analysis also revealed that among human GBM samples, EZH2 expression was homogeneous; in contrast, the heterogeneous expression of estrogen receptors (ERs) allowed the classification of the samples into groups. Even in the GBM cluster with high expression of ERs and those of their target genes, the expression of PCR2 target genes did not change. Overall, our data suggest that in GBM cells, pro-oncogenic actions of E2 are mediated by EZH2, without changes in EZH2 expression and by mechanisms that appear to be unrelated to the transcriptional activity of ERs.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste , Glioblastoma , Movimento Celular/genética , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Estradiol/farmacologia , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos
3.
Exp Hematol ; 85: 57-69, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32437908

RESUMO

Inhibition of the H3K79 histone methyltransferase DOT1L has exhibited encouraging preclinical and early clinical activity in KMT2A (MLL)-rearranged leukemia, supporting the development of combinatorial therapies. Here, we investigated two novel combinations: dual inhibition of the histone methyltransferases DOT1L and EZH2, and the combination with a protein synthesis inhibitor. EZH2 is the catalytic subunit in the polycomb repressive complex 2 (PRC2), and inhibition of EZH2 has been reported to have preclinical activity in KMT2A-r leukemia. When combined with DOT1L inhibition, however, we observed both synergistic and antagonistic effects. Interestingly, antagonistic effects were not due to PRC2-mediated de-repression of HOXA9. HOXA cluster genes are key canonical targets of both KMT2A and the PRC2 complex. The independence of the HOXA cluster from PRC2 repression in KMT2A-r leukemia thus affords important insights into leukemia biology. Further studies revealed that EZH2 inhibition counteracted the effect of DOT1L inhibition on ribosomal gene expression. We thus identified a previously unrecognized role of DOT1L in regulating protein production. Decreased translation was one of the earliest effects measurable after DOT1L inhibition and specific to KMT2A-rearranged cell lines. H3K79me2 chromatin immunoprecipitation sequencing patterns over ribosomal genes were similar to those of the canonical KMT2A-fusion target genes in primary AML patient samples. The effects of DOT1L inhibition on ribosomal gene expression prompted us to evaluate the combination of EPZ5676 with a protein translation inhibitor. EPZ5676 was synergistic with the protein translation inhibitor homoharringtonine (omacetaxine), supporting further preclinical/clinical development of this combination. In summary, we discovered a novel epigenetic regulation of a metabolic process-protein synthesis-that plays a role in leukemogenesis and affords a combinatorial therapeutic opportunity.


Assuntos
Epigênese Genética , Regulação Leucêmica da Expressão Gênica , Rearranjo Gênico , Leucemia Mieloide Aguda/metabolismo , Biossíntese de Proteínas , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/genética , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo
4.
Exp Mol Pathol ; 113: 104374, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31917966

RESUMO

The endocrine disruptive effects caused by bisphenol A (BPA) are well known. Despite this, to date, evaluation of its long term effects is limited, meaning that there is still much to be unveiled in terms of alterations caused by perinatal exposure to BPA. Our aim was to determine if perinatal exposure to two different doses of BPA causes long term morphological and molecular alteration effects in the mammary gland (MG). We evaluated MG from Mongolian gerbil offspring exposed perinatally (during gestation and lactation) to 50 or 5000 µg/kg/day BPA. At 90 days of age the animals were subjected to a single dose of N-nitroso-N-methylurea in order to mimic a carcinogenic environment. At 6 months of age, animals in estrous were euthanized for morphological evaluation of the MGs. The MG architecture presented considerable changes in terms of detached epithelial cells, inflammation, glandular hyperplasia, and collagen fiber deposition. Furthermore, a higher index of epithelial cell proliferation was detected in comparison to the intact control group. In addition, we verified a higher molecular expression of EZH2 in the vehicle treated group, indicating that corn oil applied alone can alter the expression of this epigenetic biomarker. In conclusion, BPA perinatal exposure promotes significant changes in glandular cytoarchitecture and increases glandular epithelium proliferation rate, leading to the retention of stem-like properties. This event could compromise the fate and differentiation potential of mammary epithelium.


Assuntos
Envelhecimento/patologia , Compostos Benzidrílicos/toxicidade , Glândulas Mamárias Animais/patologia , Fenóis/toxicidade , Efeitos Tardios da Exposição Pré-Natal/patologia , Actinas/metabolismo , Animais , Proliferação de Células , Colágeno/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Gerbillinae , Histonas/metabolismo , Glândulas Mamárias Animais/efeitos dos fármacos , Gravidez
5.
Oncol Rep ; 42(3): 1248-1256, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31322273

RESUMO

Ribavirin exhibits inhibitory effects on the epigenetic enzyme enhancer of zeste homolog 2 (EZH2), which participates in lymphomagenesis. Additionally, preclinical and clinical studies have demonstrated the anti­lymphoma activity of this drug. To further investigate the potential of ribavirin as an anticancer treatment for lymphoma, the tumor­suppressive effects of ribavirin were analyzed in lymphoma cell lines. The effects of ribavirin on the viability and clonogenicity of the B­cell lymphoma cell line Pfeiffer (EZH2­mutant), Toledo (EZH2 wild­type) and cutaneous T­cell lymphoma Hut78 cell line were assessed. Expression of EZH2 and trimethylation status of histone 3, lysine 27 trimethylated (H3K27m3) was also determined in response to ribavirin. The transcriptional effects of ribavirin on Hut78 cells were analyzed by microarray expression and the results were validated by reverse transcription­quantitative polymerase chain reaction, western blotting and knockout of signal transducer and activator of transcription 1 (STAT1). The results of the present study demonstrated that ribavirin suppressed the growth and clonogenicity of cells in a dose­dependent manner. Ribavirin did not affect the expression of EZH2 nor altered its activity as evaluated by H3K27 trimethylation status. Furthermore, the results of transcriptome analysis indicated that the majority of the canonical pathways affected by ribavirin were associated with the immune system, including 'antigen presentation', 'communication between innate and adaptive immune cells' and 'cross­talk between dendritic and natural killer cells'. The results of gene expression analysis were confirmed, by demonstrating at the RNA and protein levels, downregulation of stearoyl­CoA desaturase and upregulation of STAT1. Depletion of STAT1, which was proposed as a key regulator of the aforementioned pathways, exerted growth inhibitory effects almost to the same extent as ribavirin. In conclusion, ribavirin was proposed to exert growth inhibitory effects on lymphoma cell lines, particularly Hut78 cells, a cutaneous T­cell lymphoma cell line. Of note, these effects may depend on, at least in part, the activation of canonical immune pathways regulated by the key factors STAT1 and interferon­Î³. Our results provide insight into the anti­lymphoma potential of ribavirin; however, further investigations in preclinical and clinical studies are required to determine the effectiveness of ribavirin as a therapeutic agent for treating lymphoma.


Assuntos
Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfoma/patologia , Ribavirina/farmacologia , Biomarcadores Tumorais/metabolismo , Metilação de DNA , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigenômica , Perfilação da Expressão Gênica , Humanos , Linfoma/tratamento farmacológico , Linfoma/genética , Linfoma/metabolismo , Células Tumorais Cultivadas
6.
Genet Mol Res ; 15(2)2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27323178

RESUMO

We investigated the expression and clinical implications of enhancer of Zeste homolog 2 (EZH2) and p53 protein in cervical squamous cell carcinoma (SCC) and precancerous lesions. EZH2 and p53 expressions in SCC (168), cervical intraepithelial neoplasia (CIN)-I (19), CIN-II (35), and normal tissues (30) were detected by streptavidin-peroxidase-conjugation. The correlation between co-expression of EZH2 and p53 protein and the clinic pathological features and prognosis of SCC were discussed. The positive expression rates of EZH2 and p53 were 6.7, 37.0, and 75.6%, and 3.3, 21.1, and 39.3% in normal cervical tissues, CIN, and SCC, respectively, which were significantly different (P < 0.05). The positive expression rate of EZH2 and p53 protein in SCC patients with and without lymph node metastasis was 82.9 and 70.4% (EZH2) and 45.7 and 34.7% (p53), respectively, which was also a significant difference (P < 0.05). The progression-free survival (PFS) rates in followed-up patients (N = 143) who were EZH2- and p53-negative, EZH2- or p53-positive, and EZH2- and p53-positive were 71.3 ± 1.9, 66.1 ± 2.0, and 51.3 ± 3.8 months, respectively, which was a significant difference (P < 0.001); the overall survival among these groups was 72.9 ± 1.1, 68.6 ± 1.8, and 57.4 ± 3.4 months, respectively (P < 0.001). Multivariate analyses revealed that EZH2 expression, lymph node metastasis, and tumor staging were independent prognostic factors of SCC. EZH2 and p53, which affect lymph node metastasis and prognosis of SCC, may play a key role in the occurrence and development of SCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Displasia do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/metabolismo , Adulto , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Intervalo Livre de Doença , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Displasia do Colo do Útero/genética , Displasia do Colo do Útero/patologia
7.
Genet Mol Res ; 15(1)2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-27050973

RESUMO

We determined whether the coexpression of Notch1 and EZH2 influences the progression and prognosis of breast invasive ductal carcinoma. Using the χ(2) test, a significant difference was found between high and low expression of Notch1 in terms of lymph node, hormone receptor, and p53 expression (P < 0.05). Moreover, a significant difference was found between high and low expression of EZH2 in terms of tumor size, histologic grade, hormone receptor, and expression of Ki67 (P < 0.05). Using Pearson correlation analysis, we found a significant positive correlation between Notch1 and EZH2 expression in the tissue samples of breast invasive ductal carcinoma (P = 0.038). High Notch1 and EZH2 expression was associated with poor progression-free survival compared with low expression (PNotch1 = 0.000, 40.3 vs 48.9 months; PEZH2 = 0.000, 40.2 vs 49.9 months). Moreover, we found that high Notch1 and EZH2 expression was associated with poor overall survival compared with low expression (PNotch1 = 0.000, 51.2 vs 56.2 months; PEZH2 = 0.002, 51.7 vs 56.4 months). In conclusion, Notch1 and EZH2 coexpression contributes to the progression and prognosis of breast invasive ductal carcinoma.


Assuntos
Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Receptor Notch1/metabolismo , Adulto , Idoso , Carcinoma Ductal de Mama/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Pessoa de Meia-Idade , Prognóstico , Receptor Notch1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA