Cargando…

Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility

KRAS plays critical roles in regulating a range of normal cellular events as well as pathological processes in many tissues mediated through a variety of signaling pathways, including ERK1/2 and AKT signaling, in a cell-, context- and development-dependent manner. The in vivo function of KRAS and it...

Descripción completa

Detalles Bibliográficos
Autores principales: Sun, Penghao, Wang, Hongliang, Liu, Lingyun, Guo, Kaimin, Li, Xian, Cao, Yin, Ko, Chemyong, Lan, Zi-Jian, Lei, Zhenmin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9437434/
https://www.ncbi.nlm.nih.gov/pubmed/36060690
http://dx.doi.org/10.3389/fphys.2022.991719
_version_ 1784781610255646720
author Sun, Penghao
Wang, Hongliang
Liu, Lingyun
Guo, Kaimin
Li, Xian
Cao, Yin
Ko, Chemyong
Lan, Zi-Jian
Lei, Zhenmin
author_facet Sun, Penghao
Wang, Hongliang
Liu, Lingyun
Guo, Kaimin
Li, Xian
Cao, Yin
Ko, Chemyong
Lan, Zi-Jian
Lei, Zhenmin
author_sort Sun, Penghao
collection PubMed
description KRAS plays critical roles in regulating a range of normal cellular events as well as pathological processes in many tissues mediated through a variety of signaling pathways, including ERK1/2 and AKT signaling, in a cell-, context- and development-dependent manner. The in vivo function of KRAS and its downstream targets in gonadal steroidogenic cells for the development and homeostasis of reproductive functions remain to be determined. To understand the functions of KRAS signaling in gonadal theca and interstitial cells, we generated a Kras mutant (tKrasMT) mouse line that selectively expressed a constitutively active Kras ( G12D ) in these cells. Kras ( G12D ) expression in ovarian theca cells did not block follicle development to the preovulatory stage. However, tKrasMT females failed to ovulate and thus were infertile. The phosphorylated ERK1/2 and forkhead box O1 (FOXO1) and total FOXO1 protein levels were markedly reduced in tKrasMT theca cells. Kras ( G12D ) expression in theca cells also curtailed the phosphorylation of ERK1/2 and altered the expression of several ovulation-related genes in gonadotropin-primed granulosa cells. To uncover downstream targets of KRAS/FOXO1 signaling in theca cells, we found that the expression of bone morphogenic protein 7 (Bmp7), a theca-specific factor involved in ovulation, was significantly elevated in tKrasMT theca cells. Chromosome immunoprecipitation assays demonstrated that FOXO1 interacted with the Bmp7 promoter containing forkhead response elements and that the binding activity was attenuated in tKrasMT theca cells. Moreover, Foxo1 knockdown caused an elevation, whereas Foxo1 overexpression resulted in an inhibition of Bmp7 expression, suggesting that KRAS signaling regulates FOXO1 protein levels to control Bmp7 expression in theca cells. Thus, the anovulation phenotype observed in tKrasMT mice may be attributed to aberrant KRAS/FOXO1/BMP7 signaling in theca cells. Our work provides the first in vivo evidence that maintaining normal KRAS activity in ovarian theca cells is crucial for ovulation and female fertility.
format Online
Article
Text
id pubmed-9437434
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-94374342022-09-03 Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility Sun, Penghao Wang, Hongliang Liu, Lingyun Guo, Kaimin Li, Xian Cao, Yin Ko, Chemyong Lan, Zi-Jian Lei, Zhenmin Front Physiol Physiology KRAS plays critical roles in regulating a range of normal cellular events as well as pathological processes in many tissues mediated through a variety of signaling pathways, including ERK1/2 and AKT signaling, in a cell-, context- and development-dependent manner. The in vivo function of KRAS and its downstream targets in gonadal steroidogenic cells for the development and homeostasis of reproductive functions remain to be determined. To understand the functions of KRAS signaling in gonadal theca and interstitial cells, we generated a Kras mutant (tKrasMT) mouse line that selectively expressed a constitutively active Kras ( G12D ) in these cells. Kras ( G12D ) expression in ovarian theca cells did not block follicle development to the preovulatory stage. However, tKrasMT females failed to ovulate and thus were infertile. The phosphorylated ERK1/2 and forkhead box O1 (FOXO1) and total FOXO1 protein levels were markedly reduced in tKrasMT theca cells. Kras ( G12D ) expression in theca cells also curtailed the phosphorylation of ERK1/2 and altered the expression of several ovulation-related genes in gonadotropin-primed granulosa cells. To uncover downstream targets of KRAS/FOXO1 signaling in theca cells, we found that the expression of bone morphogenic protein 7 (Bmp7), a theca-specific factor involved in ovulation, was significantly elevated in tKrasMT theca cells. Chromosome immunoprecipitation assays demonstrated that FOXO1 interacted with the Bmp7 promoter containing forkhead response elements and that the binding activity was attenuated in tKrasMT theca cells. Moreover, Foxo1 knockdown caused an elevation, whereas Foxo1 overexpression resulted in an inhibition of Bmp7 expression, suggesting that KRAS signaling regulates FOXO1 protein levels to control Bmp7 expression in theca cells. Thus, the anovulation phenotype observed in tKrasMT mice may be attributed to aberrant KRAS/FOXO1/BMP7 signaling in theca cells. Our work provides the first in vivo evidence that maintaining normal KRAS activity in ovarian theca cells is crucial for ovulation and female fertility. Frontiers Media S.A. 2022-08-19 /pmc/articles/PMC9437434/ /pubmed/36060690 http://dx.doi.org/10.3389/fphys.2022.991719 Text en Copyright © 2022 Sun, Wang, Liu, Guo, Li, Cao, Ko, Lan and Lei. 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 Physiology
Sun, Penghao
Wang, Hongliang
Liu, Lingyun
Guo, Kaimin
Li, Xian
Cao, Yin
Ko, Chemyong
Lan, Zi-Jian
Lei, Zhenmin
Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility
title Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility
title_full Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility
title_fullStr Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility
title_full_unstemmed Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility
title_short Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility
title_sort aberrant activation of kras in mouse theca-interstitial cells results in female infertility
topic Physiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9437434/
https://www.ncbi.nlm.nih.gov/pubmed/36060690
http://dx.doi.org/10.3389/fphys.2022.991719
work_keys_str_mv AT sunpenghao aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT wanghongliang aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT liulingyun aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT guokaimin aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT lixian aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT caoyin aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT kochemyong aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT lanzijian aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility
AT leizhenmin aberrantactivationofkrasinmousethecainterstitialcellsresultsinfemaleinfertility