Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites. / Guérillon, Claire; Smedegaard, Stine; Hendriks, Ivo A; Nielsen, Michael L; Mailand, Niels.

In: Journal of Biological Chemistry, Vol. 295, 2020, p. 8350-8362.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Guérillon, C, Smedegaard, S, Hendriks, IA, Nielsen, ML & Mailand, N 2020, 'Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites', Journal of Biological Chemistry, vol. 295, pp. 8350-8362. https://doi.org/10.1074/jbc.RA120.013780

APA

Guérillon, C., Smedegaard, S., Hendriks, I. A., Nielsen, M. L., & Mailand, N. (2020). Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites. Journal of Biological Chemistry, 295, 8350-8362. https://doi.org/10.1074/jbc.RA120.013780

Vancouver

Guérillon C, Smedegaard S, Hendriks IA, Nielsen ML, Mailand N. Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites. Journal of Biological Chemistry. 2020;295:8350-8362. https://doi.org/10.1074/jbc.RA120.013780

Author

Guérillon, Claire ; Smedegaard, Stine ; Hendriks, Ivo A ; Nielsen, Michael L ; Mailand, Niels. / Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites. In: Journal of Biological Chemistry. 2020 ; Vol. 295. pp. 8350-8362.

Bibtex

@article{bd6fb173239a4371897030c6cf586afc,
title = "Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites",
abstract = "Translesion DNA synthesis (TLS) mediated by low-fidelity DNA polymerases is an essential cellular mechanism for bypassing DNA lesions that obstruct DNA replication progression. However, the access of TLS polymerases to the replication machinery must be kept tightly in check in order to avoid excessive mutagenesis. Recruitment of DNA polymerase η (Pol η) and other Y-family TLS polymerases to damaged DNA relies on proliferating cell nuclear antigen (PCNA) monoubiquitylation and is regulated at several levels. Using a microscopy-based RNAi screen, here we identified an important role of the SUMO modification pathway in limiting Pol η interactions with DNA damage sites in human cells. We found that Pol η undergoes DNA damage- and protein inhibitor of activated STAT 1 (PIAS1)-dependent polySUMOylation upon its association with monoubiquitylated PCNA, rendering it susceptible to extraction from DNA damage sites by SUMO-targeted ubiquitin ligase (STUbL) activity. Using proteomic profiling, we demonstrate that Pol η is targeted for multi-site SUMOylation, and that collectively these SUMO modifications are essential for PIAS1- and STUbL-mediated displacement of Pol η from DNA damage sites. These findings suggest that a SUMO-driven feedback inhibition mechanism is an intrinsic feature of TLS-mediated lesion bypass functioning to curtail the interaction of Pol η with PCNA at damaged DNA to prevent harmful mutagenesis.",
author = "Claire Gu{\'e}rillon and Stine Smedegaard and Hendriks, {Ivo A} and Nielsen, {Michael L} and Niels Mailand",
year = "2020",
doi = "10.1074/jbc.RA120.013780",
language = "English",
volume = "295",
pages = "8350--8362",
journal = "Journal of Biological Chemistry",
issn = "0021-9258",
publisher = "American Society for Biochemistry and Molecular Biology, Inc.",

}

RIS

TY - JOUR

T1 - Multisite SUMOylation restrains DNA polymerase η interactions with DNA damage sites

AU - Guérillon, Claire

AU - Smedegaard, Stine

AU - Hendriks, Ivo A

AU - Nielsen, Michael L

AU - Mailand, Niels

PY - 2020

Y1 - 2020

N2 - Translesion DNA synthesis (TLS) mediated by low-fidelity DNA polymerases is an essential cellular mechanism for bypassing DNA lesions that obstruct DNA replication progression. However, the access of TLS polymerases to the replication machinery must be kept tightly in check in order to avoid excessive mutagenesis. Recruitment of DNA polymerase η (Pol η) and other Y-family TLS polymerases to damaged DNA relies on proliferating cell nuclear antigen (PCNA) monoubiquitylation and is regulated at several levels. Using a microscopy-based RNAi screen, here we identified an important role of the SUMO modification pathway in limiting Pol η interactions with DNA damage sites in human cells. We found that Pol η undergoes DNA damage- and protein inhibitor of activated STAT 1 (PIAS1)-dependent polySUMOylation upon its association with monoubiquitylated PCNA, rendering it susceptible to extraction from DNA damage sites by SUMO-targeted ubiquitin ligase (STUbL) activity. Using proteomic profiling, we demonstrate that Pol η is targeted for multi-site SUMOylation, and that collectively these SUMO modifications are essential for PIAS1- and STUbL-mediated displacement of Pol η from DNA damage sites. These findings suggest that a SUMO-driven feedback inhibition mechanism is an intrinsic feature of TLS-mediated lesion bypass functioning to curtail the interaction of Pol η with PCNA at damaged DNA to prevent harmful mutagenesis.

AB - Translesion DNA synthesis (TLS) mediated by low-fidelity DNA polymerases is an essential cellular mechanism for bypassing DNA lesions that obstruct DNA replication progression. However, the access of TLS polymerases to the replication machinery must be kept tightly in check in order to avoid excessive mutagenesis. Recruitment of DNA polymerase η (Pol η) and other Y-family TLS polymerases to damaged DNA relies on proliferating cell nuclear antigen (PCNA) monoubiquitylation and is regulated at several levels. Using a microscopy-based RNAi screen, here we identified an important role of the SUMO modification pathway in limiting Pol η interactions with DNA damage sites in human cells. We found that Pol η undergoes DNA damage- and protein inhibitor of activated STAT 1 (PIAS1)-dependent polySUMOylation upon its association with monoubiquitylated PCNA, rendering it susceptible to extraction from DNA damage sites by SUMO-targeted ubiquitin ligase (STUbL) activity. Using proteomic profiling, we demonstrate that Pol η is targeted for multi-site SUMOylation, and that collectively these SUMO modifications are essential for PIAS1- and STUbL-mediated displacement of Pol η from DNA damage sites. These findings suggest that a SUMO-driven feedback inhibition mechanism is an intrinsic feature of TLS-mediated lesion bypass functioning to curtail the interaction of Pol η with PCNA at damaged DNA to prevent harmful mutagenesis.

U2 - 10.1074/jbc.RA120.013780

DO - 10.1074/jbc.RA120.013780

M3 - Journal article

C2 - 32350109

VL - 295

SP - 8350

EP - 8362

JO - Journal of Biological Chemistry

JF - Journal of Biological Chemistry

SN - 0021-9258

ER -

ID: 240408809