Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.

Regulation of poly(ADP-ribose) (PAR) synthesis and turnover is critical to determining cell fate after genotoxic stress. Hyperactivation of PAR synthesis by poly(ADP-ribose) polymerase-1 (PARP-1) occurs when cells deficient in DNA repair are exposed to genotoxic agents; however, the function of this...

Full description

Bibliographic Details
Main Authors: Natalie R Gassman, Donna F Stefanick, Padmini S Kedar, Julie K Horton, Samuel H Wilson
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2012-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3492265?pdf=render
id doaj-5097241639b2458cb11e13c5211c71e8
record_format Article
spelling doaj-5097241639b2458cb11e13c5211c71e82020-11-24T20:50:00ZengPublic Library of Science (PLoS)PLoS ONE1932-62032012-01-01711e4930110.1371/journal.pone.0049301Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.Natalie R GassmanDonna F StefanickPadmini S KedarJulie K HortonSamuel H WilsonRegulation of poly(ADP-ribose) (PAR) synthesis and turnover is critical to determining cell fate after genotoxic stress. Hyperactivation of PAR synthesis by poly(ADP-ribose) polymerase-1 (PARP-1) occurs when cells deficient in DNA repair are exposed to genotoxic agents; however, the function of this hyperactivation has not been adequately explained. Here, we examine PAR synthesis in mouse fibroblasts deficient in the base excision repair enzyme DNA polymerase β (pol β). The extent and duration of PARP-1 activation was measured after exposure to either the DNA alkylating agent, methyl methanesulfonate (MMS), or to low energy laser-induced DNA damage. There was strong DNA damage-induced hyperactivation of PARP-1 in pol β nullcells, but not in wild-type cells. In the case of MMS treatment, PAR synthesis did not lead to cell death in the pol β null cells, but instead resulted in increased PARylation of the nonhomologous end-joining (NHEJ) protein Ku70 and increased association of Ku70 with PARP-1. Inhibition of the NHEJ factor DNA-PK, under conditions of MMS-induced PARP-1 hyperactivation, enhanced necrotic cell death. These data suggest that PARP-1 hyperactivation is a protective mechanism triggering the classical-NHEJ DNA repair pathway when the primary alkylated base damage repair pathway is compromised.http://europepmc.org/articles/PMC3492265?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Natalie R Gassman
Donna F Stefanick
Padmini S Kedar
Julie K Horton
Samuel H Wilson
spellingShingle Natalie R Gassman
Donna F Stefanick
Padmini S Kedar
Julie K Horton
Samuel H Wilson
Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
PLoS ONE
author_facet Natalie R Gassman
Donna F Stefanick
Padmini S Kedar
Julie K Horton
Samuel H Wilson
author_sort Natalie R Gassman
title Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
title_short Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
title_full Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
title_fullStr Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
title_full_unstemmed Hyperactivation of PARP triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
title_sort hyperactivation of parp triggers nonhomologous end-joining in repair-deficient mouse fibroblasts.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2012-01-01
description Regulation of poly(ADP-ribose) (PAR) synthesis and turnover is critical to determining cell fate after genotoxic stress. Hyperactivation of PAR synthesis by poly(ADP-ribose) polymerase-1 (PARP-1) occurs when cells deficient in DNA repair are exposed to genotoxic agents; however, the function of this hyperactivation has not been adequately explained. Here, we examine PAR synthesis in mouse fibroblasts deficient in the base excision repair enzyme DNA polymerase β (pol β). The extent and duration of PARP-1 activation was measured after exposure to either the DNA alkylating agent, methyl methanesulfonate (MMS), or to low energy laser-induced DNA damage. There was strong DNA damage-induced hyperactivation of PARP-1 in pol β nullcells, but not in wild-type cells. In the case of MMS treatment, PAR synthesis did not lead to cell death in the pol β null cells, but instead resulted in increased PARylation of the nonhomologous end-joining (NHEJ) protein Ku70 and increased association of Ku70 with PARP-1. Inhibition of the NHEJ factor DNA-PK, under conditions of MMS-induced PARP-1 hyperactivation, enhanced necrotic cell death. These data suggest that PARP-1 hyperactivation is a protective mechanism triggering the classical-NHEJ DNA repair pathway when the primary alkylated base damage repair pathway is compromised.
url http://europepmc.org/articles/PMC3492265?pdf=render
work_keys_str_mv AT nataliergassman hyperactivationofparptriggersnonhomologousendjoininginrepairdeficientmousefibroblasts
AT donnafstefanick hyperactivationofparptriggersnonhomologousendjoininginrepairdeficientmousefibroblasts
AT padminiskedar hyperactivationofparptriggersnonhomologousendjoininginrepairdeficientmousefibroblasts
AT juliekhorton hyperactivationofparptriggersnonhomologousendjoininginrepairdeficientmousefibroblasts
AT samuelhwilson hyperactivationofparptriggersnonhomologousendjoininginrepairdeficientmousefibroblasts
_version_ 1716805072263839744