RAD51AP1-deficiency in vertebrate cells impairs DNA replication

Standard

RAD51AP1-deficiency in vertebrate cells impairs DNA replication. / Parplys, Ann C; Kratz, Katja; Speed, Michael C; Leung, Stanley G; Schild, David; Wiese, Claudia.

In: DNA REPAIR, Vol. 24, 05.10.2014, p. 87-97.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Parplys, AC, Kratz, K, Speed, MC, Leung, SG, Schild, D & Wiese, C 2014, 'RAD51AP1-deficiency in vertebrate cells impairs DNA replication', DNA REPAIR, vol. 24, pp. 87-97. https://doi.org/10.1016/j.dnarep.2014.09.007

APA

Parplys, A. C., Kratz, K., Speed, M. C., Leung, S. G., Schild, D., & Wiese, C. (2014). RAD51AP1-deficiency in vertebrate cells impairs DNA replication. DNA REPAIR, 24, 87-97. https://doi.org/10.1016/j.dnarep.2014.09.007

Vancouver

Parplys AC, Kratz K, Speed MC, Leung SG, Schild D, Wiese C. RAD51AP1-deficiency in vertebrate cells impairs DNA replication. DNA REPAIR. 2014 Oct 5;24:87-97. https://doi.org/10.1016/j.dnarep.2014.09.007

Bibtex

@article{117af377bfec498b8345195501845f5b,
title = "RAD51AP1-deficiency in vertebrate cells impairs DNA replication",
abstract = "RAD51-associated protein 1 (RAD51AP1) is critical for homologous recombination (HR) by interacting with and stimulating the activities of the RAD51 and DMC1 recombinases. In human somatic cells, knockdown of RAD51AP1 results in increased sensitivity to DNA damaging agents and to impaired HR, but the formation of DNA damage-induced RAD51 foci is unaffected. Here, we generated a genetic model system, based on chicken DT40 cells, to assess the phenotype of fully inactivated RAD51AP1 in vertebrate cells. Targeted inactivation of both RAD51AP1 alleles has no effect on either viability or doubling-time in undamaged cells, but leads to increased levels of cytotoxicity after exposure to cisplatin or to ionizing radiation. Interestingly, ectopic expression of GgRAD51AP1, but not of HsRAD51AP1 is able to fully complement in cell survival assays. Notably, in RAD51AP1-deficient DT40 cells the resolution of DNA damage-induced RAD51 foci is greatly slowed down, while their formation is not impaired. We also identify, for the first time, an important role for RAD51AP1 in counteracting both spontaneous and DNA damage-induced replication stress. In human and in chicken cells, RAD51AP1 is required to maintain wild type speed of replication fork progression, and both RAD51AP1-depleted human cells and RAD51AP1-deficient DT40 cells respond to replication stress by a slow-down of replication fork elongation rates. However, increased firing of replication origins occurs in RAD51AP1-/- DT40 cells, likely to ensure the timely duplication of the entire genome. Taken together, our results may explain why RAD51AP1 commonly is overexpressed in tumor cells and tissues, and we speculate that the disruption of RAD51AP1 function could be a promising approach in targeted tumor therapy.",
author = "Parplys, {Ann C} and Katja Kratz and Speed, {Michael C} and Leung, {Stanley G} and David Schild and Claudia Wiese",
note = "Copyright {\textcopyright} 2014 Elsevier B.V. All rights reserved.",
year = "2014",
month = oct,
day = "5",
doi = "10.1016/j.dnarep.2014.09.007",
language = "English",
volume = "24",
pages = "87--97",
journal = "DNA REPAIR",
issn = "1568-7864",
publisher = "Elsevier",

}

RIS

TY - JOUR

T1 - RAD51AP1-deficiency in vertebrate cells impairs DNA replication

AU - Parplys, Ann C

AU - Kratz, Katja

AU - Speed, Michael C

AU - Leung, Stanley G

AU - Schild, David

AU - Wiese, Claudia

N1 - Copyright © 2014 Elsevier B.V. All rights reserved.

PY - 2014/10/5

Y1 - 2014/10/5

N2 - RAD51-associated protein 1 (RAD51AP1) is critical for homologous recombination (HR) by interacting with and stimulating the activities of the RAD51 and DMC1 recombinases. In human somatic cells, knockdown of RAD51AP1 results in increased sensitivity to DNA damaging agents and to impaired HR, but the formation of DNA damage-induced RAD51 foci is unaffected. Here, we generated a genetic model system, based on chicken DT40 cells, to assess the phenotype of fully inactivated RAD51AP1 in vertebrate cells. Targeted inactivation of both RAD51AP1 alleles has no effect on either viability or doubling-time in undamaged cells, but leads to increased levels of cytotoxicity after exposure to cisplatin or to ionizing radiation. Interestingly, ectopic expression of GgRAD51AP1, but not of HsRAD51AP1 is able to fully complement in cell survival assays. Notably, in RAD51AP1-deficient DT40 cells the resolution of DNA damage-induced RAD51 foci is greatly slowed down, while their formation is not impaired. We also identify, for the first time, an important role for RAD51AP1 in counteracting both spontaneous and DNA damage-induced replication stress. In human and in chicken cells, RAD51AP1 is required to maintain wild type speed of replication fork progression, and both RAD51AP1-depleted human cells and RAD51AP1-deficient DT40 cells respond to replication stress by a slow-down of replication fork elongation rates. However, increased firing of replication origins occurs in RAD51AP1-/- DT40 cells, likely to ensure the timely duplication of the entire genome. Taken together, our results may explain why RAD51AP1 commonly is overexpressed in tumor cells and tissues, and we speculate that the disruption of RAD51AP1 function could be a promising approach in targeted tumor therapy.

AB - RAD51-associated protein 1 (RAD51AP1) is critical for homologous recombination (HR) by interacting with and stimulating the activities of the RAD51 and DMC1 recombinases. In human somatic cells, knockdown of RAD51AP1 results in increased sensitivity to DNA damaging agents and to impaired HR, but the formation of DNA damage-induced RAD51 foci is unaffected. Here, we generated a genetic model system, based on chicken DT40 cells, to assess the phenotype of fully inactivated RAD51AP1 in vertebrate cells. Targeted inactivation of both RAD51AP1 alleles has no effect on either viability or doubling-time in undamaged cells, but leads to increased levels of cytotoxicity after exposure to cisplatin or to ionizing radiation. Interestingly, ectopic expression of GgRAD51AP1, but not of HsRAD51AP1 is able to fully complement in cell survival assays. Notably, in RAD51AP1-deficient DT40 cells the resolution of DNA damage-induced RAD51 foci is greatly slowed down, while their formation is not impaired. We also identify, for the first time, an important role for RAD51AP1 in counteracting both spontaneous and DNA damage-induced replication stress. In human and in chicken cells, RAD51AP1 is required to maintain wild type speed of replication fork progression, and both RAD51AP1-depleted human cells and RAD51AP1-deficient DT40 cells respond to replication stress by a slow-down of replication fork elongation rates. However, increased firing of replication origins occurs in RAD51AP1-/- DT40 cells, likely to ensure the timely duplication of the entire genome. Taken together, our results may explain why RAD51AP1 commonly is overexpressed in tumor cells and tissues, and we speculate that the disruption of RAD51AP1 function could be a promising approach in targeted tumor therapy.

U2 - 10.1016/j.dnarep.2014.09.007

DO - 10.1016/j.dnarep.2014.09.007

M3 - SCORING: Journal article

C2 - 25288561

VL - 24

SP - 87

EP - 97

JO - DNA REPAIR

JF - DNA REPAIR

SN - 1568-7864

ER -