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.
PLoS Genet ; 10(9): e1004642, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25255445

RESUMO

There are two major pathways leading to induction of NF-κB subunits. The classical (or canonical) pathway typically leads to the induction of RelA or c-Rel containing complexes, and involves the degradation of IκBα in a manner dependent on IκB kinase (IKK) ß and the IKK regulatory subunit NEMO. The alternative (or non-canonical) pathway, involves the inducible processing of p100 to p52, leading to the induction of NF-κB2(p52)/RelB containing complexes, and is dependent on IKKα and NF-κB inducing kinase (NIK). Here we demonstrate that in primary human fibroblasts, the alternative NF-κB pathway subunits NF-κB2 and RelB have multiple, but distinct, effects on the expression of key regulators of the cell cycle, reactive oxygen species (ROS) generation and protein stability. Specifically, following siRNA knockdown, quantitative PCR, western blot analyses and chromatin immunoprecipitation (ChIP) show that NF-κB2 regulates the expression of CDK4 and CDK6, while RelB, through the regulation of genes such as PSMA5 and ANAPC1, regulates the stability of p21WAF1 and the tumour suppressor p53. These combine to regulate the activity of the retinoblastoma protein, Rb, leading to induction of polycomb protein EZH2 expression. Moreover, our ChIP analysis demonstrates that EZH2 is also a direct NF-κB target gene. Microarray analysis revealed that in fibroblasts, EZH2 antagonizes a subset of p53 target genes previously associated with the senescent cell phenotype, including DEK and RacGAP1. We show that this pathway provides the major route of crosstalk between the alternative NF-κB pathway and p53, a consequence of which is to suppress cell senescence. Importantly, we find that activation of NF-κB also induces EZH2 expression in CD40L stimulated cells from Chronic Lymphocytic Leukemia patients. We therefore propose that this pathway provides a mechanism through which microenvironment induced NF-κB can inhibit tumor suppressor function and promote tumorigenesis.


Assuntos
Senescência Celular/genética , NF-kappa B/metabolismo , Complexo Repressor Polycomb 2/genética , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Ligante de CD40/agonistas , Ligante de CD40/metabolismo , Análise por Conglomerados , Proteína Potenciadora do Homólogo 2 de Zeste , Ativação Enzimática , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/metabolismo , Modelos Biológicos , Subunidade p52 de NF-kappa B/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica , Estabilidade Proteica , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição RelB/metabolismo , Transcrição Gênica , Transcriptoma
2.
Biochem Soc Trans ; 42(1): 76-81, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24450631

RESUMO

The NF-κB (nuclear factor κB) transcription factor family is a pleiotropic regulator of many cellular pathways, providing a mechanism for the cell to respond to a wide variety of stimuli and environmental challenges. It is not surprising therefore that an important component of NF-κB's function includes regulation of the cell cycle. However, this aspect of its behaviour is often overlooked and receives less attention than its ability to induce inflammatory gene expression. In the present article, we provide an updated review of the current state of our knowledge about integration of NF-κB activity with cell cycle regulation, including newly characterized direct and indirect target genes in addition to the mechanisms through which NF-κB itself can be regulated by the cell cycle.


Assuntos
Mitose , NF-kappa B/fisiologia , Animais , Ciclina D1/genética , Ciclina D1/metabolismo , Regulação da Expressão Gênica , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional , Transcrição Gênica
3.
Cell Cycle ; 12(18): 3052-62, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23974100

RESUMO

Activation of the NFκB signaling pathway allows the cell to respond to infection and stress and can affect many cellular processes. As a consequence, NFκB activity must be integrated with a wide variety of parallel signaling pathways. One mechanism through which NFκB can exert widespread effects is through controlling the expression of key regulatory kinases. Here we report that NFκB regulates the expression of genes required for centrosome duplication, and that Polo-like kinase 4 (PLK4) is a direct NFκB target gene. RNA interference, chromatin immunoprecipitation, and analysis of the PLK4 promoter in a luciferase reporter assay revealed that all NFκB subunits participate in its regulation. Moreover, we demonstrate that NFκB regulation of PLK4 expression is seen in multiple cell types. Significantly long-term deletion of the NFκB2 (p100/p52) subunit leads to defects in centrosome structure. This data reveals a new component of cell cycle regulation by NFκB and suggests a mechanism through which deregulated NFκB activity in cancer can lead to increased genomic instability and uncontrolled proliferation.


Assuntos
Subunidade p52 de NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Linhagem Celular Tumoral , Centrossomo/química , Centrossomo/metabolismo , Imunoprecipitação da Cromatina , Instabilidade Genômica , Células HeLa , Humanos , Subunidade p52 de NF-kappa B/antagonistas & inibidores , Subunidade p52 de NF-kappa B/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...