Cargando…

Bmi1 Deficient Mice Exhibit Male Infertility

Previous studies have demonstrated that the polycomb repressor Bmi1 is universally expressed in all types of testicular cells and might regulate the spermatogonia proliferation, however, it is unclear whether Bmi1 plays a critical role in maintaining normal male fertility in vivo. To answer this que...

Descripción completa

Detalles Bibliográficos
Autores principales: Dai, Xiuliang, Zhang, Qian, Yu, Zhenzhen, Sun, Weiwei, Wang, Rong, Miao, Dengshun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5859480/
https://www.ncbi.nlm.nih.gov/pubmed/29559852
http://dx.doi.org/10.7150/ijbs.23325
_version_ 1783307830991781888
author Dai, Xiuliang
Zhang, Qian
Yu, Zhenzhen
Sun, Weiwei
Wang, Rong
Miao, Dengshun
author_facet Dai, Xiuliang
Zhang, Qian
Yu, Zhenzhen
Sun, Weiwei
Wang, Rong
Miao, Dengshun
author_sort Dai, Xiuliang
collection PubMed
description Previous studies have demonstrated that the polycomb repressor Bmi1 is universally expressed in all types of testicular cells and might regulate the spermatogonia proliferation, however, it is unclear whether Bmi1 plays a critical role in maintaining normal male fertility in vivo. To answer this question, we first confirmed that Bmi1 is universally expressed in all types of testicular cells and found that the gene relative expression levels of Bmi1 in testis were the highest relative to other organs. Then we investigated the role of Bmi1 in maintaining normal male fertility using Bmi1 knockout male mouse model. Our results demonstrated that Bmi1 deficiency resulted in totally male infertility with smaller testis, severe oligospermia and sperm malformation. Mechanistically, decreased serum testosterone levels with down-regulating 3βHSD and 17βHSD expression levels, reduced germ cell proliferation, increased germ cell apoptosis with up-regulating p16, p19, p53 and p21 expression levels, increased reactive oxygen species (ROS) and H(2)O(2) levels with down-regulating gene expression levels of anti-oxidant enzymes, and increased 8-OHdG and γ.H2AX positive cells in testis were observed in Bmi1 deficient mice compared with wild-type mice. These results indicate that Bmi1 deficiency results in male infertility by reducing testosterone syntheses, increasing oxidative stress and DNA damage, activating p16 and p19 signaling pathway, inhibiting germ cell proliferation and inducing germ cell apoptosis and sperm malformation. Thus, Bmi1 may be a novel and potential target for the clinic treatment of male infertility.
format Online
Article
Text
id pubmed-5859480
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-58594802018-03-20 Bmi1 Deficient Mice Exhibit Male Infertility Dai, Xiuliang Zhang, Qian Yu, Zhenzhen Sun, Weiwei Wang, Rong Miao, Dengshun Int J Biol Sci Research Paper Previous studies have demonstrated that the polycomb repressor Bmi1 is universally expressed in all types of testicular cells and might regulate the spermatogonia proliferation, however, it is unclear whether Bmi1 plays a critical role in maintaining normal male fertility in vivo. To answer this question, we first confirmed that Bmi1 is universally expressed in all types of testicular cells and found that the gene relative expression levels of Bmi1 in testis were the highest relative to other organs. Then we investigated the role of Bmi1 in maintaining normal male fertility using Bmi1 knockout male mouse model. Our results demonstrated that Bmi1 deficiency resulted in totally male infertility with smaller testis, severe oligospermia and sperm malformation. Mechanistically, decreased serum testosterone levels with down-regulating 3βHSD and 17βHSD expression levels, reduced germ cell proliferation, increased germ cell apoptosis with up-regulating p16, p19, p53 and p21 expression levels, increased reactive oxygen species (ROS) and H(2)O(2) levels with down-regulating gene expression levels of anti-oxidant enzymes, and increased 8-OHdG and γ.H2AX positive cells in testis were observed in Bmi1 deficient mice compared with wild-type mice. These results indicate that Bmi1 deficiency results in male infertility by reducing testosterone syntheses, increasing oxidative stress and DNA damage, activating p16 and p19 signaling pathway, inhibiting germ cell proliferation and inducing germ cell apoptosis and sperm malformation. Thus, Bmi1 may be a novel and potential target for the clinic treatment of male infertility. Ivyspring International Publisher 2018-03-09 /pmc/articles/PMC5859480/ /pubmed/29559852 http://dx.doi.org/10.7150/ijbs.23325 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Dai, Xiuliang
Zhang, Qian
Yu, Zhenzhen
Sun, Weiwei
Wang, Rong
Miao, Dengshun
Bmi1 Deficient Mice Exhibit Male Infertility
title Bmi1 Deficient Mice Exhibit Male Infertility
title_full Bmi1 Deficient Mice Exhibit Male Infertility
title_fullStr Bmi1 Deficient Mice Exhibit Male Infertility
title_full_unstemmed Bmi1 Deficient Mice Exhibit Male Infertility
title_short Bmi1 Deficient Mice Exhibit Male Infertility
title_sort bmi1 deficient mice exhibit male infertility
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5859480/
https://www.ncbi.nlm.nih.gov/pubmed/29559852
http://dx.doi.org/10.7150/ijbs.23325
work_keys_str_mv AT daixiuliang bmi1deficientmiceexhibitmaleinfertility
AT zhangqian bmi1deficientmiceexhibitmaleinfertility
AT yuzhenzhen bmi1deficientmiceexhibitmaleinfertility
AT sunweiwei bmi1deficientmiceexhibitmaleinfertility
AT wangrong bmi1deficientmiceexhibitmaleinfertility
AT miaodengshun bmi1deficientmiceexhibitmaleinfertility