Cargando…

PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes

BACKGROUND: Intrauterine adhesion (IUA) is a reproductive dysfunction disease characterized by endometrial fibrosis, with limited therapeutic options and poor prognosis. Our previous studies confirmed that menstrual blood-derived stromal cells (MenSCs) effectively attenuated endometrial fibrosis in...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Xudong, Zhang, Siwen, Qi, Jiarui, Zhao, Fujie, Lu, Yimeng, Li, Shuyu, Wu, Shanshan, Li, Pingping, Tan, Jichun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10148410/
https://www.ncbi.nlm.nih.gov/pubmed/37118830
http://dx.doi.org/10.1186/s13287-023-03339-y
_version_ 1785034969313181696
author Zhang, Xudong
Zhang, Siwen
Qi, Jiarui
Zhao, Fujie
Lu, Yimeng
Li, Shuyu
Wu, Shanshan
Li, Pingping
Tan, Jichun
author_facet Zhang, Xudong
Zhang, Siwen
Qi, Jiarui
Zhao, Fujie
Lu, Yimeng
Li, Shuyu
Wu, Shanshan
Li, Pingping
Tan, Jichun
author_sort Zhang, Xudong
collection PubMed
description BACKGROUND: Intrauterine adhesion (IUA) is a reproductive dysfunction disease characterized by endometrial fibrosis, with limited therapeutic options and poor prognosis. Our previous studies confirmed that menstrual blood-derived stromal cells (MenSCs) effectively attenuated endometrial fibrosis in an animal model of IUA mainly through exosomes. This therapeutic effect can be enhanced by platelet-rich plasma (PRP), in which PDGFBB is an abundant growth factor. Therefore, we aimed to compare the effects of PRP and PDGFBB on the biological activities of MenSCs in vitro, and to further investigate the molecular mechanism of MenSCs-derived exosomes in alleviating endometrial fibrosis. METHODS: MenSCs were isolated for in vitro functional assays to examine the viability, migration, and stemness of MenSCs. Endometrial stromal cells (EndoSCs) were treated with 50 ug/ml of MenSCs-derived exosomes, obtained by differential ultracentrifugation extraction. The molecular mechanisms by which PDGFBB improves MenSCs and exosomes alleviate EndoSCs fibrosis were then explored using immunofluorescence, western blot, and co-immunoprecipitation. RESULTS: Both 100 ng/ml PDGFBB and 10% activated PRP promoted the proliferation, increased the S phase of cell cycle, and inhibited apoptosis of MenSCs in vitro. Compared with PRP, PDGFBB significantly promoted MenSCs migration. All of these effects were inhibited by sorafenib, a PDGFR-β inhibitor. PRP and PDGFBB activated AKT/NF-κB signaling pathway in MenSCs and increased the expression of P65 and OCT4. Moreover, pretreatment of PDGFBB did not increase the secretion of MenSCs but significantly increased the anti-fibrosis effects of MenSCs-derived exosomes on IUA-EndoSCs. MenSCs-derived exosomes attenuated SMAD3 phosphorylation and increased YAP ubiquitination, which reduced the binding of YAP/SMAD3. Pretreatment with PDGFBB amplified this effect. CONCLUSIONS: In summary, PDGFBB could improve the biological functions of MenSCs via AKT/NF-κB signaling pathway, including viability, migration, and stemness. Our results indicated that PDGFBB amplified MenSCs-derived exosomes to attenuate endometrial fibrosis by inhibiting YAP activity, revealing a novel mechanism by which PRP enhanced the ability of MenSCs to repair tissue injury and providing a potential option for improving stem cell efficacy in IUA. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-023-03339-y.
format Online
Article
Text
id pubmed-10148410
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-101484102023-04-30 PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes Zhang, Xudong Zhang, Siwen Qi, Jiarui Zhao, Fujie Lu, Yimeng Li, Shuyu Wu, Shanshan Li, Pingping Tan, Jichun Stem Cell Res Ther Research BACKGROUND: Intrauterine adhesion (IUA) is a reproductive dysfunction disease characterized by endometrial fibrosis, with limited therapeutic options and poor prognosis. Our previous studies confirmed that menstrual blood-derived stromal cells (MenSCs) effectively attenuated endometrial fibrosis in an animal model of IUA mainly through exosomes. This therapeutic effect can be enhanced by platelet-rich plasma (PRP), in which PDGFBB is an abundant growth factor. Therefore, we aimed to compare the effects of PRP and PDGFBB on the biological activities of MenSCs in vitro, and to further investigate the molecular mechanism of MenSCs-derived exosomes in alleviating endometrial fibrosis. METHODS: MenSCs were isolated for in vitro functional assays to examine the viability, migration, and stemness of MenSCs. Endometrial stromal cells (EndoSCs) were treated with 50 ug/ml of MenSCs-derived exosomes, obtained by differential ultracentrifugation extraction. The molecular mechanisms by which PDGFBB improves MenSCs and exosomes alleviate EndoSCs fibrosis were then explored using immunofluorescence, western blot, and co-immunoprecipitation. RESULTS: Both 100 ng/ml PDGFBB and 10% activated PRP promoted the proliferation, increased the S phase of cell cycle, and inhibited apoptosis of MenSCs in vitro. Compared with PRP, PDGFBB significantly promoted MenSCs migration. All of these effects were inhibited by sorafenib, a PDGFR-β inhibitor. PRP and PDGFBB activated AKT/NF-κB signaling pathway in MenSCs and increased the expression of P65 and OCT4. Moreover, pretreatment of PDGFBB did not increase the secretion of MenSCs but significantly increased the anti-fibrosis effects of MenSCs-derived exosomes on IUA-EndoSCs. MenSCs-derived exosomes attenuated SMAD3 phosphorylation and increased YAP ubiquitination, which reduced the binding of YAP/SMAD3. Pretreatment with PDGFBB amplified this effect. CONCLUSIONS: In summary, PDGFBB could improve the biological functions of MenSCs via AKT/NF-κB signaling pathway, including viability, migration, and stemness. Our results indicated that PDGFBB amplified MenSCs-derived exosomes to attenuate endometrial fibrosis by inhibiting YAP activity, revealing a novel mechanism by which PRP enhanced the ability of MenSCs to repair tissue injury and providing a potential option for improving stem cell efficacy in IUA. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-023-03339-y. BioMed Central 2023-04-28 /pmc/articles/PMC10148410/ /pubmed/37118830 http://dx.doi.org/10.1186/s13287-023-03339-y Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Zhang, Xudong
Zhang, Siwen
Qi, Jiarui
Zhao, Fujie
Lu, Yimeng
Li, Shuyu
Wu, Shanshan
Li, Pingping
Tan, Jichun
PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
title PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
title_full PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
title_fullStr PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
title_full_unstemmed PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
title_short PDGFBB improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
title_sort pdgfbb improved the biological function of menstrual blood-derived stromal cells and the anti-fibrotic properties of exosomes
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10148410/
https://www.ncbi.nlm.nih.gov/pubmed/37118830
http://dx.doi.org/10.1186/s13287-023-03339-y
work_keys_str_mv AT zhangxudong pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT zhangsiwen pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT qijiarui pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT zhaofujie pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT luyimeng pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT lishuyu pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT wushanshan pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT lipingping pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes
AT tanjichun pdgfbbimprovedthebiologicalfunctionofmenstrualbloodderivedstromalcellsandtheantifibroticpropertiesofexosomes