Abstract

Estrogens can elicit rapid cellular responses via the G-protein-coupled receptor 30 (GPR30), followed by estrogen receptor α (ERα/ESR1)-mediated genomic effects. Here, we investigated whether rapid estrogen signaling via GRP30 may affect ESR1 expression, and we examined the underlying molecular mechanisms. The exposure of human endometrial cancer cells to 17β-estradiol promoted p62 phosphorylation and increased ESR1 protein expression. However, both a GPR30 antagonist and GPR30 silencing abrogated this phenomenon. GPR30 activation by 17β-estradiol elicited the SRC/EGFR/PI3K/Akt/mTOR signaling pathway. Intriguingly, unphosphorylated p62 and ESR1 were found to form an intracellular complex with the substrate adaptor protein KEAP1. Upon phosphorylation, p62 promoted ESR1 release from the complex, to increase its protein expression. Given the critical role played by p62 in autophagy, we also examined how this process affected ESR1 expression. The activation of autophagy by everolimus decreased ESR1 by promoting p62 degradation, whereas autophagy inhibition with chloroquine increased ESR1 expression. The treatment of female C57BL/6 mice with the autophagy inhibitor hydroxychloroquine—which promotes p62 expression—increased both phosphorylated p62 and ESR1 expression in uterine epithelial cells. Collectively, our results indicate that 17β-estradiol-mediated GPR30 activation elicits the SRC/EGFR/PI3K/Akt/mTOR signaling pathway and promotes p62 phosphorylation. In turn, phosphorylated p62 increased ESR1 expression by inducing its release from complexes that included KEAP1. Our findings may lead to novel pharmacological strategies aimed at decreasing ESR1 expression in estrogen-sensitive cells.

Highlights

  • While estrogens play key roles in the female reproductive system and fertility, abnormal exposure has long been considered a significant risk factor for endometrial and breast malignancies [1]

  • We sought to shed light on the molecular underpinnings of 17β-estradiol signaling through G-protein-coupled receptor 30 (GPR30)—with a special focus on the roles played by p62 and KEAP1 as potential mediators. Knowledge of this complex interplay may pave the way for novel signaling- or autophagy-mediated pharmacological strategies aimed at decreasing ESR1 expression in estrogen-sensitive cells

  • The results of our study indicate that GPR30 activation by 17β-estradiol elicits the SRC/EGFR/PI3K/Akt/mTOR signaling pathway and promotes p62 phosphorylation—

Read more

Summary

Introduction

While estrogens play key roles in the female reproductive system and fertility, abnormal exposure has long been considered a significant risk factor for endometrial and breast malignancies [1]. Estrogens are known to exert canonical genomic effects that occur mainly via the activation of estrogen receptor α (ESR1) [2]. Estrogens are capable of inducing rapid alterations in signaling processes [3], such as effects mediated by specific interactions with the G-protein-coupled receptor 30 (GPR30), to activate the PI3K/AKT and EGFR/MAPK signaling pathways [4]. Other studies have demonstrated an adverse prognostic impact of GRP30 overexpression in gynecological malignancies [7,8]. While both conventional effects of estrogens are well attested, evidence to support the idea that rapid signaling via GRP30 possesses the ability to modulate canonical estrogen genomic signaling, by influencing

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call