Your browser doesn't support javascript.
loading
Loss of lamin B receptor is necessary to induce cellular senescence.
Lukásová, Emilie; Kovarˇík, Ales; Bacˇíková, Alena; Falk, Martin; Kozubek, Stanislav.
Afiliação
  • Lukásová E; Department of Cell Biology and Radiobiology, Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, Brno 61265, Czech Republic lukasova@ibp.cz.
  • Kovarˇík A; Department of Molecular Epigenetics, Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, Brno 61265, Czech Republic.
  • Bacˇíková A; Department of Cell Biology and Radiobiology, Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, Brno 61265, Czech Republic.
  • Falk M; Department of Cell Biology and Radiobiology, Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, Brno 61265, Czech Republic.
  • Kozubek S; Department of Cell Biology and Radiobiology, Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, Brno 61265, Czech Republic.
Biochem J ; 474(2): 281-300, 2017 01 15.
Article em En | MEDLINE | ID: mdl-27760841
ABSTRACT
Cellular transition to senescence is associated with extensive chromatin reorganization and changes in gene expression. Recent studies appear to imply an association of lamin B1 (LB1) reduction with chromatin rearrangement in human fibroblasts promoted to senescence, while the mechanisms and structural features of these relationships have not yet been clarified. In this work, we examined the functions of LB1 and the lamin B receptor (LBR) in human cancer cells. We found that both LB1 and LBR tend to deplete during cancer cell transfer to senescence by γ-irradiation. A functional study employing silencing of LBR by small hairpin ribonucleic acid (shRNA) constructs revealed reduced LB1 levels suggesting that the regulation of both proteins is interrelated. The reduced expression of LBR resulted in the relocation of centromeric heterochromatin (CSH) from the inner nuclear membrane (INM) to the nucleoplasm and is associated with its unfolding. This indicates that LBR tethers heterochromatin to INM in cycling cancer cells and that LB1 is an integral part of this tethering. Down-regulation of LBR and LB1 at the onset of senescence are thus necessary for the release of heterochromatin binding to lamina, resulting in changes in chromatin architecture and gene expression. However, the senescence phenotype was not manifested in cell lines with reduced LBR and LB1 expression suggesting that other factors, such as deoxyribonucleic acid (DNA) damage, are needed to trigger senescence. We conclude that the primary response of cells to various stresses leading to senescence consists of the down-regulation of LBR and LB1 to attain reversal of the chromatin architecture.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Osteoblastos / Heterocromatina / Regulação Neoplásica da Expressão Gênica / Receptores Citoplasmáticos e Nucleares / Lamina Tipo B Limite: Humans Idioma: En Revista: Biochem J Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Osteoblastos / Heterocromatina / Regulação Neoplásica da Expressão Gênica / Receptores Citoplasmáticos e Nucleares / Lamina Tipo B Limite: Humans Idioma: En Revista: Biochem J Ano de publicação: 2017 Tipo de documento: Article