Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 130(11): 1952-1964, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28446540

RESUMO

Many pathways dysregulated in prostate cancer are also involved in epithelial differentiation. To better understand prostate tumor initiation, we sought to investigate specific genes and mechanisms required for normal basal to luminal cell differentiation. Utilizing human prostate basal epithelial cells and an in vitro differentiation model, we tested the hypothesis that regulation of NOTCH3 by the p38 MAPK family (hereafter p38-MAPK), via MYC, is required for luminal differentiation. Inhibition (SB202190 and BIRB796) or knockdown of p38α (also known as MAPK14) and/or p38δ (also known as MAPK13) prevented proper differentiation. Additionally, treatment with a γ-secretase inhibitor (RO4929097) or knockdown of NOTCH1 and/or NOTCH3 greatly impaired differentiation and caused luminal cell death. Constitutive p38-MAPK activation through MKK6(CA) increased NOTCH3 (but not NOTCH1) mRNA and protein levels, which was diminished upon MYC inhibition (10058-F4 and JQ1) or knockdown. Furthermore, we validated two NOTCH3 enhancer elements through a combination of enhancer (e)RNA detection (BruUV-seq) and luciferase reporter assays. Finally, we found that the NOTCH3 mRNA half-life increased during differentiation or upon acute p38-MAPK activation. These results reveal a new connection between p38-MAPK, MYC and NOTCH signaling, demonstrate two mechanisms of NOTCH3 regulation and provide evidence for NOTCH3 involvement in prostate luminal cell differentiation.


Assuntos
Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-myc/genética , Receptor Notch3/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Diferenciação Celular , Linhagem Celular Transformada , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Genes Reporter , Células HEK293 , Meia-Vida , Humanos , Imidazóis/farmacologia , Luciferases/genética , Luciferases/metabolismo , Masculino , Naftalenos/farmacologia , Cultura Primária de Células , Próstata/citologia , Próstata/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor Notch3/antagonistas & inibidores , Receptor Notch3/metabolismo , Transdução de Sinais , Tiazóis/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Prostate ; 77(1): 49-59, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27527891

RESUMO

BACKGROUND: How prostate epithelial cells differentiate and how dysregulation of this process contributes to prostate tumorigenesis remain unclear. We recently identified a Myc target and chromatin reader protein, ING4, as a necessary component of human prostate luminal epithelial cell differentiation, which is often lost in primary prostate tumors. Furthermore, loss of ING4 in the context of oncogenic mutations is required for prostate tumorigenesis. Identifying the gene targets of ING4 can provide insight into how its loss disrupts differentiation and leads to prostate cancer. METHODS: Using a combination of RNA-Seq, a best candidate approach, and chromatin immunoprecipitation (ChIP), we identified Miz1 as a new ING4 target. ING4 or Miz1 overexpression, shRNA knock-down, and a Myc-binding mutant were used in a human in vitro differentiation assay to assess the role of Miz1 in luminal cell differentiation. RESULTS: ING4 directly binds the Miz1 promoter and is required to induce Miz1 mRNA and protein expression during luminal cell differentiation. Miz1 mRNA was not induced in shING4 expressing cells or tumorigenic cells in which ING4 is not expressed. Miz1 dependency on ING4 was unique to differentiating luminal cells; Miz1 mRNA expression was not induced in basal cells. Although Miz1 is a direct target of ING4, and its overexpression can drive luminal cell differentiation, Miz1 was not required for differentiation. CONCLUSIONS: Miz1 is a newly identified ING4-induced target gene which can drive prostate luminal epithelial cell differentiation although it is not absolutely required. Prostate 77:49-59, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Diferenciação Celular/fisiologia , Células Epiteliais/metabolismo , Proteínas de Homeodomínio/biossíntese , Fatores de Transcrição Kruppel-Like/biossíntese , Próstata/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Células HEK293 , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Próstata/citologia , Ligação Proteica/fisiologia , Proteínas Supressoras de Tumor/metabolismo
3.
Cancer Res ; 74(12): 3357-68, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24762396

RESUMO

The mechanisms by which Myc overexpression or Pten loss promotes prostate cancer development are poorly understood. We identified the chromatin remodeling protein, ING4, as a crucial switch downstream of Myc and Pten that is required for human prostate epithelial differentiation. Myc-induced transient expression of ING4 is required for the differentiation of basal epithelial cells into luminal cells, while sustained ING4 expression induces apoptosis. ING4 expression is lost in >60% of human primary prostate tumors. ING4 or Pten loss prevents epithelial cell differentiation, which was necessary for tumorigenesis. Pten loss prevents differentiation by blocking ING4 expression, which is rescued by ING4 re-expression. Pten or ING4 loss generates tumor cells that co-express basal and luminal markers, indicating prostate oncogenesis occurs through disruption of an intermediate step in the prostate epithelial differentiation program. Thus, we identified a new epithelial cell differentiation switch involving Myc, Pten, and ING4, which when disrupted leads to prostate tumorigenesis. Myc overexpression and Pten loss are common genetic abnormalities in prostate cancer, whereas loss of the tumor suppressor ING4 has not been reported. This is the first demonstration that transient ING4 expression is absolutely required for epithelial differentiation, its expression is dependent on Myc and Pten, and it is lost in the majority of human prostate cancers. This is the first demonstration that loss of ING4, either directly or indirectly through loss of Pten, promotes Myc-driven oncogenesis by deregulating differentiation. The clinical implication is that Pten/ING4 negative and ING4-only negative tumors may reflect two distinct subtypes of prostate cancer.


Assuntos
Carcinogênese/metabolismo , Proteínas de Ciclo Celular/genética , Células Epiteliais/fisiologia , Proteínas de Homeodomínio/genética , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-myc/fisiologia , Ativação Transcricional , Proteínas Supressoras de Tumor/genética , Animais , Apoptose , Carcinogênese/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...