Cargando…

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

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...

Descripción completa

Detalles Bibliográficos
Autores principales: Manova, Vasilissa, Singh, Satyendra K, Iliakis, George
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3471266/
https://www.ncbi.nlm.nih.gov/pubmed/22908892
http://dx.doi.org/10.1186/2041-9414-3-4
_version_ 1782246398818779136
author Manova, Vasilissa
Singh, Satyendra K
Iliakis, George
author_facet Manova, Vasilissa
Singh, Satyendra K
Iliakis, George
author_sort Manova, Vasilissa
collection PubMed
description 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 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.
format Online
Article
Text
id pubmed-3471266
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-34712662012-10-16 Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin Manova, Vasilissa Singh, Satyendra K Iliakis, George Genome Integr Research 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 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. BioMed Central 2012-08-22 /pmc/articles/PMC3471266/ /pubmed/22908892 http://dx.doi.org/10.1186/2041-9414-3-4 Text en Copyright ©2012 Manova et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 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.
spellingShingle Research
Manova, Vasilissa
Singh, Satyendra K
Iliakis, George
Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
title Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
title_full Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
title_fullStr Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
title_full_unstemmed Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
title_short Processing of DNA double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
title_sort processing of dna double strand breaks by alternative non-homologous end-joining in hyperacetylated chromatin
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3471266/
https://www.ncbi.nlm.nih.gov/pubmed/22908892
http://dx.doi.org/10.1186/2041-9414-3-4
work_keys_str_mv AT manovavasilissa processingofdnadoublestrandbreaksbyalternativenonhomologousendjoininginhyperacetylatedchromatin
AT singhsatyendrak processingofdnadoublestrandbreaksbyalternativenonhomologousendjoininginhyperacetylatedchromatin
AT iliakisgeorge processingofdnadoublestrandbreaksbyalternativenonhomologousendjoininginhyperacetylatedchromatin