Cargando…

GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice

BACKGROUND: Female mammals have a limited reproductive lifespan determined by the size of the primordial follicle pool established perinatally. Over two thirds of fetal oocytes are abolished via programmed cell death during early folliculogenesis. However, the underlying mechanisms governing fetal o...

Descripción completa

Detalles Bibliográficos
Autores principales: Wen, Jia, Yan, Hao, He, Meina, Zhang, Tuo, Mu, Xinyi, Wang, Haibin, Zhang, Hua, Xia, Guoliang, Wang, Chao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6417224/
https://www.ncbi.nlm.nih.gov/pubmed/30866939
http://dx.doi.org/10.1186/s12915-019-0641-9
_version_ 1783403525010620416
author Wen, Jia
Yan, Hao
He, Meina
Zhang, Tuo
Mu, Xinyi
Wang, Haibin
Zhang, Hua
Xia, Guoliang
Wang, Chao
author_facet Wen, Jia
Yan, Hao
He, Meina
Zhang, Tuo
Mu, Xinyi
Wang, Haibin
Zhang, Hua
Xia, Guoliang
Wang, Chao
author_sort Wen, Jia
collection PubMed
description BACKGROUND: Female mammals have a limited reproductive lifespan determined by the size of the primordial follicle pool established perinatally. Over two thirds of fetal oocytes are abolished via programmed cell death during early folliculogenesis. However, the underlying mechanisms governing fetal oocyte attrition remain largely elusive. RESULTS: Here, we demonstrate that glycogen synthase kinase-3 beta (GSK-3β) is indispensable for fetal oocyte maintenance during meiotic prophase I in mice. In vitro inhibition of GSK-3β activity or in vivo conditional deletion of Gsk-3β in the germline led to a dramatic loss of fetal oocytes via apoptosis, which subsequently resulted in a reduced capacity of the primordial follicle pool. Inhibition of GSK-3β also impeded meiotic progression in fetal oocytes and led to a deficiency in DNA double-strand break (DSB) repair associated with premature upregulation of Tap63, the major genome guardian of the female germline, following GSK-3β inhibition in fetal ovaries. Mechanistically, we demonstrated that aberrant nuclear translocation of β-catenin was responsible for the abnormal expression of TAp63 and global fetal oocyte attrition following GSK-3β inhibition. CONCLUSIONS: In summary, GSK-3β was essential for sustaining fetal oocyte survival and folliculogenesis via fine-tuning the cytoplasmic-nuclear translocation of β-catenin, which in turn modulates timely TAp63 expression during meiotic prophase I in mice. Our study provides a perspective on the physiological regulatory role of DNA damage checkpoint signaling in fetal oocyte guardianship and female fertility. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s12915-019-0641-9) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6417224
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-64172242019-03-25 GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice Wen, Jia Yan, Hao He, Meina Zhang, Tuo Mu, Xinyi Wang, Haibin Zhang, Hua Xia, Guoliang Wang, Chao BMC Biol Research Article BACKGROUND: Female mammals have a limited reproductive lifespan determined by the size of the primordial follicle pool established perinatally. Over two thirds of fetal oocytes are abolished via programmed cell death during early folliculogenesis. However, the underlying mechanisms governing fetal oocyte attrition remain largely elusive. RESULTS: Here, we demonstrate that glycogen synthase kinase-3 beta (GSK-3β) is indispensable for fetal oocyte maintenance during meiotic prophase I in mice. In vitro inhibition of GSK-3β activity or in vivo conditional deletion of Gsk-3β in the germline led to a dramatic loss of fetal oocytes via apoptosis, which subsequently resulted in a reduced capacity of the primordial follicle pool. Inhibition of GSK-3β also impeded meiotic progression in fetal oocytes and led to a deficiency in DNA double-strand break (DSB) repair associated with premature upregulation of Tap63, the major genome guardian of the female germline, following GSK-3β inhibition in fetal ovaries. Mechanistically, we demonstrated that aberrant nuclear translocation of β-catenin was responsible for the abnormal expression of TAp63 and global fetal oocyte attrition following GSK-3β inhibition. CONCLUSIONS: In summary, GSK-3β was essential for sustaining fetal oocyte survival and folliculogenesis via fine-tuning the cytoplasmic-nuclear translocation of β-catenin, which in turn modulates timely TAp63 expression during meiotic prophase I in mice. Our study provides a perspective on the physiological regulatory role of DNA damage checkpoint signaling in fetal oocyte guardianship and female fertility. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s12915-019-0641-9) contains supplementary material, which is available to authorized users. BioMed Central 2019-03-12 /pmc/articles/PMC6417224/ /pubmed/30866939 http://dx.doi.org/10.1186/s12915-019-0641-9 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided 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 Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Wen, Jia
Yan, Hao
He, Meina
Zhang, Tuo
Mu, Xinyi
Wang, Haibin
Zhang, Hua
Xia, Guoliang
Wang, Chao
GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice
title GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice
title_full GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice
title_fullStr GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice
title_full_unstemmed GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice
title_short GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice
title_sort gsk-3β protects fetal oocytes from premature death via modulating tap63 expression in mice
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6417224/
https://www.ncbi.nlm.nih.gov/pubmed/30866939
http://dx.doi.org/10.1186/s12915-019-0641-9
work_keys_str_mv AT wenjia gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT yanhao gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT hemeina gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT zhangtuo gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT muxinyi gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT wanghaibin gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT zhanghua gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT xiaguoliang gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice
AT wangchao gsk3bprotectsfetaloocytesfromprematuredeathviamodulatingtap63expressioninmice