Cargando…

AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress

Translesion DNA synthesis (TLS) and homologous recombination (HR) cooperate during S-phase to safeguard replication forks integrity. Thus, the inhibition of TLS becomes a promising point of therapeutic intervention in HR-deficient cancers, where TLS impairment might trigger synthetic lethality (SL)....

Descripción completa

Detalles Bibliográficos
Autores principales: Villafañez, Florencia, García, Iris Alejandra, Carbajosa, Sofia, Pansa, María Florencia, Mansilla, Sabrina, Llorens, María Candelaria, Angiolini, Virginia, Guantay, Laura, Jacobs, Heinz, Madauss, Kevin P., Gloger, Israel, Gottifredi, Vanesa, Bocco, Jose Luis, Soria, Gaston
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6756059/
https://www.ncbi.nlm.nih.gov/pubmed/30705406
http://dx.doi.org/10.1038/s41388-019-0724-7
_version_ 1783453341864427520
author Villafañez, Florencia
García, Iris Alejandra
Carbajosa, Sofia
Pansa, María Florencia
Mansilla, Sabrina
Llorens, María Candelaria
Angiolini, Virginia
Guantay, Laura
Jacobs, Heinz
Madauss, Kevin P.
Gloger, Israel
Gottifredi, Vanesa
Bocco, Jose Luis
Soria, Gaston
author_facet Villafañez, Florencia
García, Iris Alejandra
Carbajosa, Sofia
Pansa, María Florencia
Mansilla, Sabrina
Llorens, María Candelaria
Angiolini, Virginia
Guantay, Laura
Jacobs, Heinz
Madauss, Kevin P.
Gloger, Israel
Gottifredi, Vanesa
Bocco, Jose Luis
Soria, Gaston
author_sort Villafañez, Florencia
collection PubMed
description Translesion DNA synthesis (TLS) and homologous recombination (HR) cooperate during S-phase to safeguard replication forks integrity. Thus, the inhibition of TLS becomes a promising point of therapeutic intervention in HR-deficient cancers, where TLS impairment might trigger synthetic lethality (SL). The main limitation to test this hypothesis is the current lack of selective pharmacological inhibitors of TLS. Herein, we developed a miniaturized screening assay to identify inhibitors of PCNA ubiquitylation, a key post-translational modification required for efficient TLS activation. After screening a library of 627 kinase inhibitors, we found that targeting the pro-survival kinase AKT leads to strong impairment of PCNA ubiquitylation. Mechanistically, we found that AKT-mediated modulation of Proliferating Cell Nuclear Antigen (PCNA) ubiquitylation after UV requires the upstream activity of DNA PKcs, without affecting PCNA ubiquitylation levels in unperturbed cells. Moreover, we confirmed that persistent AKT inhibition blocks the recruitment of TLS polymerases to sites of DNA damage and impairs DNA replication forks processivity after UV irradiation, leading to increased DNA replication stress and cell death. Remarkably, when we compared the differential survival of HR-proficient vs HR-deficient cells, we found that the combination of UV irradiation and AKT inhibition leads to robust SL induction in HR-deficient cells. We link this phenotype to AKT ability to inhibit PCNA ubiquitylation, since the targeted knockdown of PCNA E3-ligase (RAD18) and a non-ubiquitylable (PCNA K164R) knock-in model recapitulate the observed SL induction. Collectively, this work identifies AKT as a novel regulator of PCNA ubiquitylation and provides the proof-of-concept of inhibiting TLS as a therapeutic approach to selectively kill HR-deficient cells submitted to replication stress.
format Online
Article
Text
id pubmed-6756059
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-67560592019-09-24 AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress Villafañez, Florencia García, Iris Alejandra Carbajosa, Sofia Pansa, María Florencia Mansilla, Sabrina Llorens, María Candelaria Angiolini, Virginia Guantay, Laura Jacobs, Heinz Madauss, Kevin P. Gloger, Israel Gottifredi, Vanesa Bocco, Jose Luis Soria, Gaston Oncogene Article Translesion DNA synthesis (TLS) and homologous recombination (HR) cooperate during S-phase to safeguard replication forks integrity. Thus, the inhibition of TLS becomes a promising point of therapeutic intervention in HR-deficient cancers, where TLS impairment might trigger synthetic lethality (SL). The main limitation to test this hypothesis is the current lack of selective pharmacological inhibitors of TLS. Herein, we developed a miniaturized screening assay to identify inhibitors of PCNA ubiquitylation, a key post-translational modification required for efficient TLS activation. After screening a library of 627 kinase inhibitors, we found that targeting the pro-survival kinase AKT leads to strong impairment of PCNA ubiquitylation. Mechanistically, we found that AKT-mediated modulation of Proliferating Cell Nuclear Antigen (PCNA) ubiquitylation after UV requires the upstream activity of DNA PKcs, without affecting PCNA ubiquitylation levels in unperturbed cells. Moreover, we confirmed that persistent AKT inhibition blocks the recruitment of TLS polymerases to sites of DNA damage and impairs DNA replication forks processivity after UV irradiation, leading to increased DNA replication stress and cell death. Remarkably, when we compared the differential survival of HR-proficient vs HR-deficient cells, we found that the combination of UV irradiation and AKT inhibition leads to robust SL induction in HR-deficient cells. We link this phenotype to AKT ability to inhibit PCNA ubiquitylation, since the targeted knockdown of PCNA E3-ligase (RAD18) and a non-ubiquitylable (PCNA K164R) knock-in model recapitulate the observed SL induction. Collectively, this work identifies AKT as a novel regulator of PCNA ubiquitylation and provides the proof-of-concept of inhibiting TLS as a therapeutic approach to selectively kill HR-deficient cells submitted to replication stress. Nature Publishing Group UK 2019-01-31 2019 /pmc/articles/PMC6756059/ /pubmed/30705406 http://dx.doi.org/10.1038/s41388-019-0724-7 Text en © The Author(s) 2019 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Villafañez, Florencia
García, Iris Alejandra
Carbajosa, Sofia
Pansa, María Florencia
Mansilla, Sabrina
Llorens, María Candelaria
Angiolini, Virginia
Guantay, Laura
Jacobs, Heinz
Madauss, Kevin P.
Gloger, Israel
Gottifredi, Vanesa
Bocco, Jose Luis
Soria, Gaston
AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
title AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
title_full AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
title_fullStr AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
title_full_unstemmed AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
title_short AKT inhibition impairs PCNA ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
title_sort akt inhibition impairs pcna ubiquitylation and triggers synthetic lethality in homologous recombination-deficient cells submitted to replication stress
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6756059/
https://www.ncbi.nlm.nih.gov/pubmed/30705406
http://dx.doi.org/10.1038/s41388-019-0724-7
work_keys_str_mv AT villafanezflorencia aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT garciairisalejandra aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT carbajosasofia aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT pansamariaflorencia aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT mansillasabrina aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT llorensmariacandelaria aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT angiolinivirginia aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT guantaylaura aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT jacobsheinz aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT madausskevinp aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT glogerisrael aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT gottifredivanesa aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT boccojoseluis aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress
AT soriagaston aktinhibitionimpairspcnaubiquitylationandtriggerssyntheticlethalityinhomologousrecombinationdeficientcellssubmittedtoreplicationstress