SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.

Impediments in replication fork progression cause genomic instability, mutagenesis, and severe pathologies. At stalled forks, RPA-coated single-stranded DNA (ssDNA) activates the ATR kinase and directs fork remodeling, 2 key early events of the replication stress response. RFWD3, a recently describe...

Full description

Bibliographic Details
Main Authors: Maïlyn Yates, Isabelle Marois, Edlie St-Hilaire, Daryl A Ronato, Billel Djerir, Chloé Brochu, Théo Morin, Ian Hammond-Martel, Sari Gezzar-Dandashi, Lisa Casimir, Elliot Drobetsky, Laurent Cappadocia, Jean-Yves Masson, Hugo Wurtele, Alexandre Maréchal
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2024-03-01
Series:PLoS Biology
Online Access:https://doi.org/10.1371/journal.pbio.3002552
_version_ 1797248482222800896
author Maïlyn Yates
Isabelle Marois
Edlie St-Hilaire
Daryl A Ronato
Billel Djerir
Chloé Brochu
Théo Morin
Ian Hammond-Martel
Sari Gezzar-Dandashi
Lisa Casimir
Elliot Drobetsky
Laurent Cappadocia
Jean-Yves Masson
Hugo Wurtele
Alexandre Maréchal
author_facet Maïlyn Yates
Isabelle Marois
Edlie St-Hilaire
Daryl A Ronato
Billel Djerir
Chloé Brochu
Théo Morin
Ian Hammond-Martel
Sari Gezzar-Dandashi
Lisa Casimir
Elliot Drobetsky
Laurent Cappadocia
Jean-Yves Masson
Hugo Wurtele
Alexandre Maréchal
author_sort Maïlyn Yates
collection DOAJ
description Impediments in replication fork progression cause genomic instability, mutagenesis, and severe pathologies. At stalled forks, RPA-coated single-stranded DNA (ssDNA) activates the ATR kinase and directs fork remodeling, 2 key early events of the replication stress response. RFWD3, a recently described Fanconi anemia (FA) ubiquitin ligase, associates with RPA and promotes its ubiquitylation, facilitating late steps of homologous recombination (HR). Intriguingly, RFWD3 also regulates fork progression, restart and stability via poorly understood mechanisms. Here, we used proteomics to identify putative RFWD3 substrates during replication stress in human cells. We show that RFWD3 interacts with and ubiquitylates the SMARCAL1 DNA translocase directly in vitro and following DNA damage in vivo. SMARCAL1 ubiquitylation does not trigger its subsequent proteasomal degradation but instead disengages it from RPA thereby regulating its function at replication forks. Proper regulation of SMARCAL1 by RFWD3 at stalled forks protects them from excessive MUS81-mediated cleavage in response to UV irradiation, thereby limiting DNA replication stress. Collectively, our results identify RFWD3-mediated SMARCAL1 ubiquitylation as a novel mechanism that modulates fork remodeling to avoid genome instability triggered by aberrant fork processing.
first_indexed 2024-04-24T20:15:17Z
format Article
id doaj.art-7a96143210c043db9c56e93cd5ee4106
institution Directory Open Access Journal
issn 1544-9173
1545-7885
language English
last_indexed 2024-04-24T20:15:17Z
publishDate 2024-03-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS Biology
spelling doaj.art-7a96143210c043db9c56e93cd5ee41062024-03-23T05:30:28ZengPublic Library of Science (PLoS)PLoS Biology1544-91731545-78852024-03-01223e300255210.1371/journal.pbio.3002552SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.Maïlyn YatesIsabelle MaroisEdlie St-HilaireDaryl A RonatoBillel DjerirChloé BrochuThéo MorinIan Hammond-MartelSari Gezzar-DandashiLisa CasimirElliot DrobetskyLaurent CappadociaJean-Yves MassonHugo WurteleAlexandre MaréchalImpediments in replication fork progression cause genomic instability, mutagenesis, and severe pathologies. At stalled forks, RPA-coated single-stranded DNA (ssDNA) activates the ATR kinase and directs fork remodeling, 2 key early events of the replication stress response. RFWD3, a recently described Fanconi anemia (FA) ubiquitin ligase, associates with RPA and promotes its ubiquitylation, facilitating late steps of homologous recombination (HR). Intriguingly, RFWD3 also regulates fork progression, restart and stability via poorly understood mechanisms. Here, we used proteomics to identify putative RFWD3 substrates during replication stress in human cells. We show that RFWD3 interacts with and ubiquitylates the SMARCAL1 DNA translocase directly in vitro and following DNA damage in vivo. SMARCAL1 ubiquitylation does not trigger its subsequent proteasomal degradation but instead disengages it from RPA thereby regulating its function at replication forks. Proper regulation of SMARCAL1 by RFWD3 at stalled forks protects them from excessive MUS81-mediated cleavage in response to UV irradiation, thereby limiting DNA replication stress. Collectively, our results identify RFWD3-mediated SMARCAL1 ubiquitylation as a novel mechanism that modulates fork remodeling to avoid genome instability triggered by aberrant fork processing.https://doi.org/10.1371/journal.pbio.3002552
spellingShingle Maïlyn Yates
Isabelle Marois
Edlie St-Hilaire
Daryl A Ronato
Billel Djerir
Chloé Brochu
Théo Morin
Ian Hammond-Martel
Sari Gezzar-Dandashi
Lisa Casimir
Elliot Drobetsky
Laurent Cappadocia
Jean-Yves Masson
Hugo Wurtele
Alexandre Maréchal
SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.
PLoS Biology
title SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.
title_full SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.
title_fullStr SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.
title_full_unstemmed SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.
title_short SMARCAL1 ubiquitylation controls its association with RPA-coated ssDNA and promotes replication fork stability.
title_sort smarcal1 ubiquitylation controls its association with rpa coated ssdna and promotes replication fork stability
url https://doi.org/10.1371/journal.pbio.3002552
work_keys_str_mv AT mailynyates smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT isabellemarois smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT edliesthilaire smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT darylaronato smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT billeldjerir smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT chloebrochu smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT theomorin smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT ianhammondmartel smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT sarigezzardandashi smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT lisacasimir smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT elliotdrobetsky smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT laurentcappadocia smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT jeanyvesmasson smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT hugowurtele smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability
AT alexandremarechal smarcal1ubiquitylationcontrolsitsassociationwithrpacoatedssdnaandpromotesreplicationforkstability