Cargando…

Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway

OBJECTIVES: This study investigated whether exosomes from LPS pretreated bone marrow mesenchymal stem cells (LPS pre-MSCs) could prolong skin graft survival. METHODS: The exosomes were isolated from the supernatant of MSCs pretreated with LPS. LPS pre-Exo and rapamycin were injected via the tail vei...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, PeiYao, Wu, Panfeng, Khan, Umar Zeb, Zhou, Zekun, Sui, Xinlei, Li, Cheng, Dong, Kangkang, Liu, Yongjun, Qing, Liming, Tang, Juyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10504750/
https://www.ncbi.nlm.nih.gov/pubmed/37716974
http://dx.doi.org/10.1186/s12951-023-02087-8
_version_ 1785106795036934144
author Zhang, PeiYao
Wu, Panfeng
Khan, Umar Zeb
Zhou, Zekun
Sui, Xinlei
Li, Cheng
Dong, Kangkang
Liu, Yongjun
Qing, Liming
Tang, Juyu
author_facet Zhang, PeiYao
Wu, Panfeng
Khan, Umar Zeb
Zhou, Zekun
Sui, Xinlei
Li, Cheng
Dong, Kangkang
Liu, Yongjun
Qing, Liming
Tang, Juyu
author_sort Zhang, PeiYao
collection PubMed
description OBJECTIVES: This study investigated whether exosomes from LPS pretreated bone marrow mesenchymal stem cells (LPS pre-MSCs) could prolong skin graft survival. METHODS: The exosomes were isolated from the supernatant of MSCs pretreated with LPS. LPS pre-Exo and rapamycin were injected via the tail vein into C57BL/6 mice allografted with BALB/c skin; graft survival was observed and evaluated. The accumulation and polarization of macrophages were examined by immunohistochemistry. The differentiation of macrophages in the spleen was analyzed by flow cytometry. For in vitro, an inflammatory model was established. Specifically, bone marrow-derived macrophages (BMDMs) were isolated and cultured with LPS (100 ng/ml) for 3 h, and were further treated with LPS pre-Exo for 24 h or 48 h. The molecular signaling pathway responsible for modulating inflammation was examined by Western blotting. The expressions of downstream inflammatory cytokines were determined by Elisa, and the polarization of macrophages was analyzed by flow cytometry. RESULTS: LPS pre-Exo could better ablate inflammation compared to untreated MSC-derived exosomes (BM-Exo). These loaded factors inhibited the expressions of inflammatory factors via a negative feedback mechanism. In vivo, LPS pre-Exo significantly attenuated inflammatory infiltration, thus improving the survival of allogeneic skin graft. Flow cytometric analysis of BMDMs showed that LPS pre-Exo were involved in the regulation of macrophage polarization and immune homeostasis during inflammation. Further investigation revealed that the NF-κB/NLRP3/procaspase-1/IL-1β signaling pathway played a key role in LPS pre-Exo-mediated regulation of macrophage polarization. Inhibiting NF-κB in BMDMs could abolish the LPS-induced activation of inflammatory pathways and the polarization of M1 macrophages while increasing the proportion of M2 cells. CONCLUSION: LPS pre-Exo are able to switch the polarization of macrophages and enhance the resolution of inflammation. This type of exosomes provides an improved immunotherapeutic potential in prolonging graft survival. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-023-02087-8.
format Online
Article
Text
id pubmed-10504750
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105047502023-09-17 Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway Zhang, PeiYao Wu, Panfeng Khan, Umar Zeb Zhou, Zekun Sui, Xinlei Li, Cheng Dong, Kangkang Liu, Yongjun Qing, Liming Tang, Juyu J Nanobiotechnology Research OBJECTIVES: This study investigated whether exosomes from LPS pretreated bone marrow mesenchymal stem cells (LPS pre-MSCs) could prolong skin graft survival. METHODS: The exosomes were isolated from the supernatant of MSCs pretreated with LPS. LPS pre-Exo and rapamycin were injected via the tail vein into C57BL/6 mice allografted with BALB/c skin; graft survival was observed and evaluated. The accumulation and polarization of macrophages were examined by immunohistochemistry. The differentiation of macrophages in the spleen was analyzed by flow cytometry. For in vitro, an inflammatory model was established. Specifically, bone marrow-derived macrophages (BMDMs) were isolated and cultured with LPS (100 ng/ml) for 3 h, and were further treated with LPS pre-Exo for 24 h or 48 h. The molecular signaling pathway responsible for modulating inflammation was examined by Western blotting. The expressions of downstream inflammatory cytokines were determined by Elisa, and the polarization of macrophages was analyzed by flow cytometry. RESULTS: LPS pre-Exo could better ablate inflammation compared to untreated MSC-derived exosomes (BM-Exo). These loaded factors inhibited the expressions of inflammatory factors via a negative feedback mechanism. In vivo, LPS pre-Exo significantly attenuated inflammatory infiltration, thus improving the survival of allogeneic skin graft. Flow cytometric analysis of BMDMs showed that LPS pre-Exo were involved in the regulation of macrophage polarization and immune homeostasis during inflammation. Further investigation revealed that the NF-κB/NLRP3/procaspase-1/IL-1β signaling pathway played a key role in LPS pre-Exo-mediated regulation of macrophage polarization. Inhibiting NF-κB in BMDMs could abolish the LPS-induced activation of inflammatory pathways and the polarization of M1 macrophages while increasing the proportion of M2 cells. CONCLUSION: LPS pre-Exo are able to switch the polarization of macrophages and enhance the resolution of inflammation. This type of exosomes provides an improved immunotherapeutic potential in prolonging graft survival. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-023-02087-8. BioMed Central 2023-09-16 /pmc/articles/PMC10504750/ /pubmed/37716974 http://dx.doi.org/10.1186/s12951-023-02087-8 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This 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, PeiYao
Wu, Panfeng
Khan, Umar Zeb
Zhou, Zekun
Sui, Xinlei
Li, Cheng
Dong, Kangkang
Liu, Yongjun
Qing, Liming
Tang, Juyu
Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway
title Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway
title_full Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway
title_fullStr Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway
title_full_unstemmed Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway
title_short Exosomes derived from LPS-preconditioned bone marrow-derived MSC modulate macrophage plasticity to promote allograft survival via the NF-κB/NLRP3 signaling pathway
title_sort exosomes derived from lps-preconditioned bone marrow-derived msc modulate macrophage plasticity to promote allograft survival via the nf-κb/nlrp3 signaling pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10504750/
https://www.ncbi.nlm.nih.gov/pubmed/37716974
http://dx.doi.org/10.1186/s12951-023-02087-8
work_keys_str_mv AT zhangpeiyao exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT wupanfeng exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT khanumarzeb exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT zhouzekun exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT suixinlei exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT licheng exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT dongkangkang exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT liuyongjun exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT qingliming exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway
AT tangjuyu exosomesderivedfromlpspreconditionedbonemarrowderivedmscmodulatemacrophageplasticitytopromoteallograftsurvivalviathenfkbnlrp3signalingpathway