Cargando…

NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis

Endometriosis is an estrogen-dependent gynecological disease. The pathogenesis of endometriosis remains controversial, although it is generally accepted that the inflammatory immune response plays a crucial role in this process. Mast cells (MCs) are multifunctional innate immune cells that accumulat...

Descripción completa

Detalles Bibliográficos
Autores principales: Guo, Xinyue, Xu, Xinxin, Li, Tiantian, Yu, Qin, Wang, Jianzhang, Chen, Yichen, Ding, Shaojie, Zhu, Libo, Zou, Gen, Zhang, Xinmei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8494307/
https://www.ncbi.nlm.nih.gov/pubmed/34630429
http://dx.doi.org/10.3389/fimmu.2021.749979
_version_ 1784579282562973696
author Guo, Xinyue
Xu, Xinxin
Li, Tiantian
Yu, Qin
Wang, Jianzhang
Chen, Yichen
Ding, Shaojie
Zhu, Libo
Zou, Gen
Zhang, Xinmei
author_facet Guo, Xinyue
Xu, Xinxin
Li, Tiantian
Yu, Qin
Wang, Jianzhang
Chen, Yichen
Ding, Shaojie
Zhu, Libo
Zou, Gen
Zhang, Xinmei
author_sort Guo, Xinyue
collection PubMed
description Endometriosis is an estrogen-dependent gynecological disease. The pathogenesis of endometriosis remains controversial, although it is generally accepted that the inflammatory immune response plays a crucial role in this process. Mast cells (MCs) are multifunctional innate immune cells that accumulate in endometriotic lesions. However, the molecular mechanism by which estrogen modulates MCs in the development of endometriosis is not well understood. Here we report that estrogen can induce the expression of NOD-like receptor family pyrin domain containing 3 (NLRP3) through estrogen receptor (ER)-α via the estrogen responsive element (ERE) in MCs. Such transcriptional regulation is necessary for the activation of NLRP3 inflammasome and the production of mature interleukin (IL)-1β in MCs. Targeted inhibition of NLRP3 significantly restrained lesion progression and fibrogenesis in a mouse model of endometriosis. Collectively, these findings suggest that MCs contribute to the development of endometriosis through NLRP3 inflammasome activation mediated by nuclear-initiated estrogen signaling pathway.
format Online
Article
Text
id pubmed-8494307
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-84943072021-10-07 NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis Guo, Xinyue Xu, Xinxin Li, Tiantian Yu, Qin Wang, Jianzhang Chen, Yichen Ding, Shaojie Zhu, Libo Zou, Gen Zhang, Xinmei Front Immunol Immunology Endometriosis is an estrogen-dependent gynecological disease. The pathogenesis of endometriosis remains controversial, although it is generally accepted that the inflammatory immune response plays a crucial role in this process. Mast cells (MCs) are multifunctional innate immune cells that accumulate in endometriotic lesions. However, the molecular mechanism by which estrogen modulates MCs in the development of endometriosis is not well understood. Here we report that estrogen can induce the expression of NOD-like receptor family pyrin domain containing 3 (NLRP3) through estrogen receptor (ER)-α via the estrogen responsive element (ERE) in MCs. Such transcriptional regulation is necessary for the activation of NLRP3 inflammasome and the production of mature interleukin (IL)-1β in MCs. Targeted inhibition of NLRP3 significantly restrained lesion progression and fibrogenesis in a mouse model of endometriosis. Collectively, these findings suggest that MCs contribute to the development of endometriosis through NLRP3 inflammasome activation mediated by nuclear-initiated estrogen signaling pathway. Frontiers Media S.A. 2021-09-22 /pmc/articles/PMC8494307/ /pubmed/34630429 http://dx.doi.org/10.3389/fimmu.2021.749979 Text en Copyright © 2021 Guo, Xu, Li, Yu, Wang, Chen, Ding, Zhu, Zou and Zhang 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
Guo, Xinyue
Xu, Xinxin
Li, Tiantian
Yu, Qin
Wang, Jianzhang
Chen, Yichen
Ding, Shaojie
Zhu, Libo
Zou, Gen
Zhang, Xinmei
NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis
title NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis
title_full NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis
title_fullStr NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis
title_full_unstemmed NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis
title_short NLRP3 Inflammasome Activation of Mast Cells by Estrogen via the Nuclear-Initiated Signaling Pathway Contributes to the Development of Endometriosis
title_sort nlrp3 inflammasome activation of mast cells by estrogen via the nuclear-initiated signaling pathway contributes to the development of endometriosis
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8494307/
https://www.ncbi.nlm.nih.gov/pubmed/34630429
http://dx.doi.org/10.3389/fimmu.2021.749979
work_keys_str_mv AT guoxinyue nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT xuxinxin nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT litiantian nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT yuqin nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT wangjianzhang nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT chenyichen nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT dingshaojie nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT zhulibo nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT zougen nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis
AT zhangxinmei nlrp3inflammasomeactivationofmastcellsbyestrogenviathenuclearinitiatedsignalingpathwaycontributestothedevelopmentofendometriosis