Cargando…

Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model

BACKGROUND: Intrauterine adhesion (IUA) is a condition caused due to damage or infection of the endometrium. It is characterized by continuous inflammation and following fibrosis and dysfunction. However, the current animal IUA models have several disadvantages, including complex operation, high mor...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Jingman, Pan, Yuchen, Yang, Jingjing, Wang, Jiali, Jiang, Qi, Dou, Huan, Hou, Yayi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9800892/
https://www.ncbi.nlm.nih.gov/pubmed/36591221
http://dx.doi.org/10.3389/fimmu.2022.945234
_version_ 1784861382980665344
author Li, Jingman
Pan, Yuchen
Yang, Jingjing
Wang, Jiali
Jiang, Qi
Dou, Huan
Hou, Yayi
author_facet Li, Jingman
Pan, Yuchen
Yang, Jingjing
Wang, Jiali
Jiang, Qi
Dou, Huan
Hou, Yayi
author_sort Li, Jingman
collection PubMed
description BACKGROUND: Intrauterine adhesion (IUA) is a condition caused due to damage or infection of the endometrium. It is characterized by continuous inflammation and following fibrosis and dysfunction. However, the current animal IUA models have several disadvantages, including complex operation, high mortality, and many extra distractions owing to opening of the abdominal cavity to expose the uterus. Mesenchymal stem cells (MSCs), which have been used in treatment of IUA, are heterogeneous and immunosuppressive. However, their therapeutic effect is not as good as expected. METHODS: Here, we successfully built a new murine IUA model, called electric tool-scratching IUA model, and applied it in our experiments to investigate the efficacy of tumor necrosis factor-α (TNF-α) primed MSCs (T-MSCs). In the new model, we used a self-made electric tool that can cause mechanical damage to the endometrium without opening the abdominal cavity. ELISA and histological staining analysis were performed to evaluate pathological features of IUA. qRT-PCR, flow cytometry and immunofluoresence staining were performed to detect the phenotypes of macrophages. TMT proteomics quantification and western blotting assay were performed to analyze the differentially expressed proteins of MSC exosomes. RESULTS: Based on the new IUA model, we found TNF-α pretreatment could enhance the ability of MSCs to relieve inflammation and reduce endometrium fibrosis. Mechanistically, T-MSC promoted macrophage polarization to M2 phenotype through exosomes. Subsequently, we found the expression of Galectin-1 was increased in T-MSC exosomes. Finally, we analyzed the gene expression pattern of Galectin-1 treated macrophages and found Galectin-1 promoted macrophage polarization to M2 phenotype mainly through the Jak-STAT signaling pathway. CONCLUSIONS: Our studies proposed an innovative mouse model and a better MSC treatment strategy for IUA.
format Online
Article
Text
id pubmed-9800892
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-98008922022-12-31 Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model Li, Jingman Pan, Yuchen Yang, Jingjing Wang, Jiali Jiang, Qi Dou, Huan Hou, Yayi Front Immunol Immunology BACKGROUND: Intrauterine adhesion (IUA) is a condition caused due to damage or infection of the endometrium. It is characterized by continuous inflammation and following fibrosis and dysfunction. However, the current animal IUA models have several disadvantages, including complex operation, high mortality, and many extra distractions owing to opening of the abdominal cavity to expose the uterus. Mesenchymal stem cells (MSCs), which have been used in treatment of IUA, are heterogeneous and immunosuppressive. However, their therapeutic effect is not as good as expected. METHODS: Here, we successfully built a new murine IUA model, called electric tool-scratching IUA model, and applied it in our experiments to investigate the efficacy of tumor necrosis factor-α (TNF-α) primed MSCs (T-MSCs). In the new model, we used a self-made electric tool that can cause mechanical damage to the endometrium without opening the abdominal cavity. ELISA and histological staining analysis were performed to evaluate pathological features of IUA. qRT-PCR, flow cytometry and immunofluoresence staining were performed to detect the phenotypes of macrophages. TMT proteomics quantification and western blotting assay were performed to analyze the differentially expressed proteins of MSC exosomes. RESULTS: Based on the new IUA model, we found TNF-α pretreatment could enhance the ability of MSCs to relieve inflammation and reduce endometrium fibrosis. Mechanistically, T-MSC promoted macrophage polarization to M2 phenotype through exosomes. Subsequently, we found the expression of Galectin-1 was increased in T-MSC exosomes. Finally, we analyzed the gene expression pattern of Galectin-1 treated macrophages and found Galectin-1 promoted macrophage polarization to M2 phenotype mainly through the Jak-STAT signaling pathway. CONCLUSIONS: Our studies proposed an innovative mouse model and a better MSC treatment strategy for IUA. Frontiers Media S.A. 2022-12-16 /pmc/articles/PMC9800892/ /pubmed/36591221 http://dx.doi.org/10.3389/fimmu.2022.945234 Text en Copyright © 2022 Li, Pan, Yang, Wang, Jiang, Dou and Hou https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Li, Jingman
Pan, Yuchen
Yang, Jingjing
Wang, Jiali
Jiang, Qi
Dou, Huan
Hou, Yayi
Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model
title Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model
title_full Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model
title_fullStr Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model
title_full_unstemmed Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model
title_short Tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote M2 macrophage polarization via Galectin-1 and modify intrauterine adhesion on a novel murine model
title_sort tumor necrosis factor-α-primed mesenchymal stem cell-derived exosomes promote m2 macrophage polarization via galectin-1 and modify intrauterine adhesion on a novel murine model
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9800892/
https://www.ncbi.nlm.nih.gov/pubmed/36591221
http://dx.doi.org/10.3389/fimmu.2022.945234
work_keys_str_mv AT lijingman tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel
AT panyuchen tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel
AT yangjingjing tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel
AT wangjiali tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel
AT jiangqi tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel
AT douhuan tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel
AT houyayi tumornecrosisfactoraprimedmesenchymalstemcellderivedexosomespromotem2macrophagepolarizationviagalectin1andmodifyintrauterineadhesiononanovelmurinemodel