miR-21-loaded BMSC-derived Exosomes repair ovarian function of autoimmune premature ovarian insufficiency by targeting MSX1
Yutao Yang,Lichao Tang,Yuanling Xiao,Wujia Huang,Meng Gao,Jiaxin Xie,Mingxin Yang,Yanhong Wu,Xiafei Fu
DOI: https://doi.org/10.1016/j.rbmo.2024.103815
IF: 4.567
2024-01-11
Reproductive BioMedicine Online
Abstract:Research Question Approximately 5–30% of premature ovarian insufficiency (POI) can be attributed to immune factors. However, there are no effective treatments currently available. Exosomes derived from mesenchymal stem cells exhibit stem cell properties, prevent immune rejection, and have carrier functions. In this study, we explored the molecular mechanism underlying the repair effect of miR-21-loaded Bone Marrow Mesenchymal Stem Cell-derived Exosome (miR-21-Exo) on autoimmune POI. Design CCK8 assay, flow cytometry, western blotting, qRT-PCR, and enzyme-linked immunosorbent assay (ELISA) verified the effect of miR-21-Exo on IFN-γ-induced KGN cells. qRT-PCR, western blotting, and dual-luciferase reporter gene assays verify that miR-21-Exo mediated MSX1 to regulate the Notch signaling pathway and that miR-21 interacts directly with MSX1. The effects of miR-21-Exo on ovaries were verified by monitoring the estrous cycle, ovarian HE staining, follicle count, ELISA, immunohistochemistry, TUNEL, western blotting, and qRT-PCR. Results Here Our results showed that miR-21-Exo promoted IFN-γ-induced KGN cell proliferation and hormone synthesis and inhibited apoptosis. Using dual-luciferase reporter gene assays, miR-21 and MSX1 were shown to have direct interactions. Moreover, our findings elucidated that miR-21-Exo inhibited cell apoptosis and promoted hormone synthesis by mediating MSX1 to regulate the Notch signaling pathway. miR-21-Exo restored the ovarian structure of autoimmune POI mice, promoted endocrine function and proliferation, and inhibited apoptosis and inflammation in vivo. Conclusions Our study demonstrated that miR-21-Exo regulates the MSX1-mediated Notch signaling pathway to inhibit granulosa cell apoptosis and improve hormone synthesis function, providing insight into the mechanism of molecular therapy for autoimmune POI treatment.
obstetrics & gynecology,reproductive biology