Cargando…

BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury

Emerging evidence suggests cellular senescence, as a consequence of excess DNA damage and deficient repair, to be a driver of brain dysfunction following repeated mild traumatic brain injury (rmTBI). This study aimed to further investigate the role of deficient DNA repair, specifically BRCA1-related...

Descripción completa

Detalles Bibliográficos
Autores principales: Leung, Emily, Taskina, Daria, Schwab, Nicole, Hazrati, Lili-Naz
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10450634/
https://www.ncbi.nlm.nih.gov/pubmed/37638313
http://dx.doi.org/10.3389/fnins.2023.1225226
_version_ 1785095245438910464
author Leung, Emily
Taskina, Daria
Schwab, Nicole
Hazrati, Lili-Naz
author_facet Leung, Emily
Taskina, Daria
Schwab, Nicole
Hazrati, Lili-Naz
author_sort Leung, Emily
collection PubMed
description Emerging evidence suggests cellular senescence, as a consequence of excess DNA damage and deficient repair, to be a driver of brain dysfunction following repeated mild traumatic brain injury (rmTBI). This study aimed to further investigate the role of deficient DNA repair, specifically BRCA1-related repair, on DNA damage-induced senescence. BRCA1, a repair protein involved in maintaining genomic integrity with multiple roles in the central nervous system, was previously reported to be significantly downregulated in post-mortem brains with a history of rmTBI. Here we examined the effects of impaired BRCA1-related repair on DNA damage-induced senescence and outcomes 1-week post-rmTBI using mice with a heterozygous knockout for BRCA1 in a sex-segregated manner. Altered BRCA1 repair with rmTBI resulted in altered anxiety-related behaviours in males and females using elevated zero maze and contextual fear conditioning. Evaluating molecular markers associated with DNA damage signalling and senescence-related pathways revealed sex-specific differences attributed to BRCA1, where females exhibited elevated DNA damage, impaired DNA damage signalling, and dampened senescence onset compared to males. Overall, the results from this study highlight sex-specific consequences of aberrant DNA repair on outcomes post-injury, and further support a need to develop sex-specific treatments following rmTBI.
format Online
Article
Text
id pubmed-10450634
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-104506342023-08-26 BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury Leung, Emily Taskina, Daria Schwab, Nicole Hazrati, Lili-Naz Front Neurosci Neuroscience Emerging evidence suggests cellular senescence, as a consequence of excess DNA damage and deficient repair, to be a driver of brain dysfunction following repeated mild traumatic brain injury (rmTBI). This study aimed to further investigate the role of deficient DNA repair, specifically BRCA1-related repair, on DNA damage-induced senescence. BRCA1, a repair protein involved in maintaining genomic integrity with multiple roles in the central nervous system, was previously reported to be significantly downregulated in post-mortem brains with a history of rmTBI. Here we examined the effects of impaired BRCA1-related repair on DNA damage-induced senescence and outcomes 1-week post-rmTBI using mice with a heterozygous knockout for BRCA1 in a sex-segregated manner. Altered BRCA1 repair with rmTBI resulted in altered anxiety-related behaviours in males and females using elevated zero maze and contextual fear conditioning. Evaluating molecular markers associated with DNA damage signalling and senescence-related pathways revealed sex-specific differences attributed to BRCA1, where females exhibited elevated DNA damage, impaired DNA damage signalling, and dampened senescence onset compared to males. Overall, the results from this study highlight sex-specific consequences of aberrant DNA repair on outcomes post-injury, and further support a need to develop sex-specific treatments following rmTBI. Frontiers Media S.A. 2023-08-10 /pmc/articles/PMC10450634/ /pubmed/37638313 http://dx.doi.org/10.3389/fnins.2023.1225226 Text en Copyright © 2023 Leung, Taskina, Schwab and Hazrati. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Neuroscience
Leung, Emily
Taskina, Daria
Schwab, Nicole
Hazrati, Lili-Naz
BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
title BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
title_full BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
title_fullStr BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
title_full_unstemmed BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
title_short BRCA1 heterozygosity promotes DNA damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
title_sort brca1 heterozygosity promotes dna damage-induced senescence in a sex-specific manner following repeated mild traumatic brain injury
topic Neuroscience
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10450634/
https://www.ncbi.nlm.nih.gov/pubmed/37638313
http://dx.doi.org/10.3389/fnins.2023.1225226
work_keys_str_mv AT leungemily brca1heterozygositypromotesdnadamageinducedsenescenceinasexspecificmannerfollowingrepeatedmildtraumaticbraininjury
AT taskinadaria brca1heterozygositypromotesdnadamageinducedsenescenceinasexspecificmannerfollowingrepeatedmildtraumaticbraininjury
AT schwabnicole brca1heterozygositypromotesdnadamageinducedsenescenceinasexspecificmannerfollowingrepeatedmildtraumaticbraininjury
AT hazratililinaz brca1heterozygositypromotesdnadamageinducedsenescenceinasexspecificmannerfollowingrepeatedmildtraumaticbraininjury