Loading...

Preprints

Study on the effect and mechanism of mesenchymal stem cell exosome-derived miR-320a regulating TGF-β1/Smads pathway in mice with premature ovarian insufficiency

Li R, Liu X, Liang X, Tang S, Xi H, Xiu Z, Zhang X, Wu Z, Duan Y.
Preprint from
Research Square
15 March 2024
PPR
PPR822428
Abstract

Introduction:

exosomes, natural nanoscale particles released by various cells, particularly Mesenchymal Stem Cells (MSCs), have gained attention for their therapeutic potential. MSC-exosomes, combining exosome benefits with MSC attributes, show promise in treating diverse conditions, including wound healing, neurological diseases, and cardiovascular issues. The Smad proteins, crucial in TGF-β superfamily signaling, play a key role in ovarian processes and germ line formation, with BMPs influencing primordial germ cell development. The intricate communication between TGF-β ligands, such as activins and inhibins, regulates folliculogenesis stages.

Aims:

and objective: The study aims to examine how miR-320a, produced from mesenchymal stem cell exosomes, regulates the TGF-β1/Smads pathway in mice with “premature ovarian insufficiency (POI)”.

Method:

Protocols established for the care and use of animals were adhered to. The Shanghai Laboratory Animal Research Center's mice were kept in a sterile setting. After inducing wounds on the skin, uMSC-Exo was administered in hydrogel to the area. Ultracentrifugation was used to separate exosomes from cell suspension media. Cell cycle and protein markers were analysed using flow cytometry. Both in situ hybridization and immunofluorescence were performed.

Result:

Figs. 1 to 5 exhibit wound diameter dynamics over time for Mock, uMSC, and HEK293T groups. All groups have a reduction in wound size (y-axis, centimetres) as healing progresses, but the uMSC-treated group shows the greatest reduction. Particle size and intensity statistics are shown on the right side of each figure; these may provide light on wound healing mechanisms and highlight the therapeutic potential of uMSC treatment. The study found that uMSC-Exo led to significant downregulation, while Antago-uMSC-Exo, an antagonist-treated group, exhibited upregulation, suggesting a reversal of inhibitory effects. The SMAD2 reporter analysis indicates that modified uMSC-Exo treatment decreases SMAD2-binding luciferase levels, with a statistically significant reduction in uMSC-Exo. Antago-uMSC-Exo shows an increase, though not statistically significant.

Conclusion:

This research reveals the distinct microRNAs in uMSC-Exos that suppress scar formation via exosome-mediated intercellular transfer, providing a fresh strategy for enhanced wound healing and regenerative medicine.