Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
15 pages
English

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris

Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin

-

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus
15 pages
English
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus

Description

Mammalian cells employ at least two subpathways of non-homologous end-joining for the repair of ionizing radiation induced DNA double strand breaks: The canonical DNA-PK-dependent form of non-homologous end-joining (D-NHEJ) and an alternative, slowly operating, error-prone backup pathway (B-NHEJ). In contrast to D-NHEJ, which operates with similar efficiency throughout the cell cycle, B-NHEJ operates more efficiently in G2-phase. Notably, B-NHEJ also shows strong and as of yet unexplained dependency on growth activity and is markedly compromised in serum-deprived cells, or in cells that enter the plateau-phase of growth. The molecular mechanisms underpinning this response remain unknown. Since chromatin structure or changes in chromatin structure are prime candidate-B-NHEJ-modulators, we study here the role of chromatin hyperacetylation, either by HDAC2 knockdown or treatment with the HDAC inhibitor TSA, on the repair by B-NHEJ of IR-induced DSBs. Results siRNA-mediated knockdown of HDAC2 fails to provoke histone hyperacetylation in Lig4 -/- MEFs and has no detectable effect on B-NHEJ function. Treatment with TSA that inhibits multiple HDACs causes efficient, reversible chromatin hyperacetylation in Lig4 -/- MEFs, as well as in human HCT116 Lig4 -/- cells and the human glioma cell line M059K. The IR yield of DSBs in TSA-treated cells remains similar to that of untreated cells despite the expected chromatin relaxation. In addition, chromatin hyperacetylation leaves unchanged repair of DSBs by B-NHEJ in irradiated exponentially growing, or plateau-phase cells. Notably, under the experimental conditions employed here, chromatin hyperacetylation fails to detectably modulate B-NHEJ in M059K cells as well. Conclusions In summary, the results show that chromatin acetylation or deacetylation does not affect the kinetics of alternative NHEJ in all types of cells examined both in exponentially growing and serum deprived cultures. We conclude that parameters beyond chromatin acetylation determine B-NHEJ efficiency in the plateau-phase of growth.

Sujets

Informations

Publié par
Publié le 01 janvier 2012
Nombre de lectures 4
Langue English
Poids de l'ouvrage 1 Mo

Extrait

Manovaet al. Genome Integrity2012,3:4 http://www.genomeintegrity.com/content/3/1/4
GENOME INTEGRITY
R E S E A R C HOpen Access Processing of DNA double strand breaks by alternative nonhomologous endjoining in hyperacetylated chromatin 1,2 11* Vasilissa Manova, Satyendra K Singhand George Iliakis
Abstract Background:Mammalian cells employ at least two subpathways of nonhomologous endjoining for the repair of ionizing radiation induced DNA double strand breaks: The canonical DNAPKdependent form of nonhomologous endjoining (DNHEJ) and an alternative, slowly operating, errorprone backup pathway (BNHEJ). In contrast to DNHEJ, which operates with similar efficiency throughout the cell cycle, BNHEJ operates more efficiently in G2phase. Notably, BNHEJ also shows strong and as of yet unexplained dependency on growth activity and is markedly compromised in serumdeprived cells, or in cells that enter the plateauphase of growth. The molecular mechanisms underpinning this response remain unknown. Since chromatin structure or changes in chromatin structure are prime candidateBNHEJmodulators, we study here the role of chromatin hyperacetylation, either by HDAC2knockdown or treatment with the HDAC inhibitor TSA, on the repair by BNHEJ of IRinduced DSBs. / Results:siRNAmediated knockdown ofHDAC2fails to provoke histone hyperacetylation inLig4MEFs and has no detectable effect on BNHEJ function. Treatment with TSA that inhibits multiple HDACs causes efficient, reversible / / chromatin hyperacetylation inLig4MEFs, as well as in human HCT116Lig4cells and the human glioma cell line M059K. The IR yield of DSBs in TSAtreated cells remains similar to that of untreated cells despite the expected chromatin relaxation. In addition, chromatin hyperacetylation leaves unchanged repair of DSBs by BNHEJ in irradiated exponentially growing, or plateauphase cells. Notably, under the experimental conditions employed here, chromatin hyperacetylation fails to detectably modulate BNHEJ in M059K cells as well. Conclusions:In summary, the results show that chromatin acetylation or deacetylation does not affect the kinetics of alternative NHEJ in all types of cells examined both in exponentially growing and serum deprived cultures. We conclude that parameters beyond chromatin acetylation determine BNHEJ efficiency in the plateauphase of growth. Keywords:DNA Double strand breaks (DSB), Ionizing radiation (IR), HDAC, Chromatin, Chromatin acetylation, NHEJ
Background It is commonly believed that DSBs induced in the gen ome of higher eukaryotes by widely diverse endogenous and exogenous factors and processes are mainly repaired by nonhomologous endjoining (NHEJ) [13]. The canon ical and widely investigated pathway of NHEJ (DNHEJ) starts with the binding to the generated ends of the Ku70/ Ku80 complex, which then helps recruit the DNA dependent protein kinase (DNAPK) as well as other
* Correspondence: Georg.Iliakis@ukessen.de 1 Institute of Medical Radiation Biology, University of DuisburgEssen Medical School, Hufelandstr. 55, 45122, Essen, Germany Full list of author information is available at the end of the article
factors, including the nuclease Artemis and the Lig4/ Xrcc4/XLF complex. Endjoining occurs rapidly, with only minimal processing of the DNA ends to render them liga table and limited polymerization [2]. When DNHEJ fails, locally in repair proficient cells, and globally in mutants with defects in DNHEJ components, or in cells treated with DNAPK inhibitors, an alternative form of end joining operating as backup to DNHEJ becomes activated (BNHEJ) [1,46]. BNHEJ utilizes Lig3 and Parp1 [711], but also histone H1 as a stabilizing factor [12] and BCR/Abl as a regulatory component [13,14]. Also components of the DNA endresection
© 2012 Manova et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
  • Univers Univers
  • Ebooks Ebooks
  • Livres audio Livres audio
  • Presse Presse
  • Podcasts Podcasts
  • BD BD
  • Documents Documents