Your browser doesn't support javascript.
loading
BOP1 contributes to the activation of autophagy in polycystic ovary syndrome via nucleolar stress response.
Ji, Rui; Zhang, Zhimo; Yang, Zhe; Chen, Xin; Yin, Tailang; Yang, Jing.
Affiliation
  • Ji R; Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China.
  • Zhang Z; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
  • Yang Z; Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China.
  • Chen X; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
  • Yin T; Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China.
  • Yang J; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
Cell Mol Life Sci ; 81(1): 101, 2024 Feb 27.
Article in En | MEDLINE | ID: mdl-38409361
ABSTRACT
Abnormal autophagy is one of the vital features in polycystic ovary syndrome (PCOS). However, the underlying molecular mechanisms remain unelucidated. In this study, we aimed to investigate whether Block of Proliferation 1 (BOP1) is involved in the onset of autophagy activation of granulosa cells in PCOS. Firstly, we found that BOP1 expression was significantly down-regulated in the ovaries of PCOS mice, which was associated with the development of PCOS. Next, local injection of lentiviral vectors in the ovary for the overexpression of BOP1 significantly alleviated the phenotypes of elevated androgens, disturbed estrous cycle, and abnormal follicular development in PCOS mice. Subsequently, we found that knockdown of BOP1 activated autophagy of granulosa cells in the in vitro experiments, whereas overexpression of BOP1 inhibited autophagy in both in vivo and in vitro models. Mechanistically, BOP1 knockdown triggered the nucleolus stress response, which caused RPL11 to be released from the nucleolus into the nucleoplasm and inhibited the E3 ubiquitination ligase of MDM2, thereby enhancing the stability of p53. Subsequently, P53 inhibited mTOR, thereby activating autophagy in granulosa cells. In addition, the mRNA level of BOP1 was negatively correlated with antral follicle count (AFC), body-mass index (BMI), serum androgen levels, and anti-Mullerian hormone (AMH) in patients with PCOS. In summary, our study demonstrates that BOP1 downregulation inhibits mTOR phosphorylation through activation of the p53-dependent nucleolus stress response, which subsequently contributes to aberrant autophagy in granulosa cells, revealing that BOP1 may be a key target for probing the mechanisms of PCOS.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Polycystic Ovary Syndrome Limits: Animals / Female / Humans Language: En Journal: Cell Mol Life Sci Journal subject: BIOLOGIA MOLECULAR Year: 2024 Document type: Article Affiliation country: China Country of publication: Suiza

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Polycystic Ovary Syndrome Limits: Animals / Female / Humans Language: En Journal: Cell Mol Life Sci Journal subject: BIOLOGIA MOLECULAR Year: 2024 Document type: Article Affiliation country: China Country of publication: Suiza