Your browser doesn't support javascript.
loading
Histone H3 Lysine 9 Acetylation Obstructs ATM Activation and Promotes Ionizing Radiation Sensitivity in Normal Stem Cells.
Meyer, Barbara; Fabbrizi, Maria Rita; Raj, Suyash; Zobel, Cheri L; Hallahan, Dennis E; Sharma, Girdhar G.
Afiliação
  • Meyer B; Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108, USA.
  • Fabbrizi MR; Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108, USA.
  • Raj S; Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108, USA.
  • Zobel CL; Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108, USA.
  • Hallahan DE; Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108, USA; Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO 63108, USA.
  • Sharma GG; Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108, USA; Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO 63108, USA. Electronic address: sharma@wustl.edu.
Stem Cell Reports ; 7(6): 1013-1022, 2016 12 13.
Article em En | MEDLINE | ID: mdl-27974220
ABSTRACT
Dynamic spatiotemporal modification of chromatin around DNA damage is vital for efficient DNA repair. Normal stem cells exhibit an attenuated DNA damage response (DDR), inefficient DNA repair, and high radiosensitivity. The impact of unique chromatin characteristics of stem cells in DDR regulation is not yet recognized. We demonstrate that murine embryonic stem cells (ES) display constitutively elevated acetylation of histone H3 lysine 9 (H3K9ac) and low H3K9 tri-methylation (H3K9me3). DNA damage-induced local deacetylation of H3K9 was abrogated in ES along with the subsequent H3K9me3. Depletion of H3K9ac in ES by suppression of monocytic leukemia zinc finger protein (MOZ) acetyltransferase improved ATM activation, DNA repair, diminished irradiation-induced apoptosis, and enhanced clonogenic survival. Simultaneous suppression of the H3K9 methyltransferase Suv39h1 abrogated the radioprotective effect of MOZ inhibition, suggesting that high H3K9ac promoted by MOZ in ES cells obstructs local upregulation of H3K9me3 and contributes to muted DDR and increased radiosensitivity.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Radiação Ionizante / Tolerância a Radiação / Histonas / Células-Tronco Embrionárias / Lisina Idioma: En Ano de publicação: 2016 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Radiação Ionizante / Tolerância a Radiação / Histonas / Células-Tronco Embrionárias / Lisina Idioma: En Ano de publicação: 2016 Tipo de documento: Article