Cargando…

Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells

BACKGROUND: Estrogen signaling is indispensable for muscle regeneration, yet the role of estrogen in the development of muscle inflammation, especially in the intramuscular T cell response, and the influence on the intrinsic immuno-behaviors of myofibers remain largely unknown. We investigated this...

Descripción completa

Detalles Bibliográficos
Autores principales: Liao, Zhao Hong, Huang, Tao, Xiao, Jiang Wei, Gu, Rui Cai, Ouyang, Jun, Wu, Gang, Liao, Hua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6661750/
https://www.ncbi.nlm.nih.gov/pubmed/31358063
http://dx.doi.org/10.1186/s13395-019-0205-2
_version_ 1783439517232922624
author Liao, Zhao Hong
Huang, Tao
Xiao, Jiang Wei
Gu, Rui Cai
Ouyang, Jun
Wu, Gang
Liao, Hua
author_facet Liao, Zhao Hong
Huang, Tao
Xiao, Jiang Wei
Gu, Rui Cai
Ouyang, Jun
Wu, Gang
Liao, Hua
author_sort Liao, Zhao Hong
collection PubMed
description BACKGROUND: Estrogen signaling is indispensable for muscle regeneration, yet the role of estrogen in the development of muscle inflammation, especially in the intramuscular T cell response, and the influence on the intrinsic immuno-behaviors of myofibers remain largely unknown. We investigated this issue using the mice model of cardiotoxin (CTX)-induced myoinjury, with or without estrogen level adjustment. METHODS: CTX injection i.m. (tibialis anterior, TA) was performed for preparing mice myoinjury model. Injection s.c. of 17β-estradiol (E(2)) or estrogen receptor antagonist 4-OHT, or ovariectomy (OVX), was used to change estrogen level of animal models in vivo. Serum E(2) level was evaluated by ELISA. Gene levels of estrogen receptor (ERs) and cytokines/chemokines in inflamed muscle were monitored by qPCR. Inflammatory infiltration was observed by immunofluorescence. Macrophage and T cell phenotypes were analyzed by FACS. Immunoblotting was used to assess protein levels of ERs and immunomolecules in C(2)C(12) myotubes treated with E(2) or 4-OHT, in the presence of IFN-γ. RESULTS: We monitored the increased serum E(2) level and the upregulated ERβ in regenerated myofibres after myotrauma. The absence of estrogen in vivo resulted in the more severe muscle inflammatory infiltration, involving the recruitment of monocyte/macrophage and CD4(+) T cells, and the heightened proinflammatory (M1) macrophage. Moreover, estrogen signaling loss led to Treg cells infiltration decrease, Th1 response elevation in inflamed muscle, and the markedly expression upregulation of immunomolecules in IFN-γ-stimulated C(2)C(12) myotubes in vitro. CONCLUSION: Our data suggest that estrogen is a positive intervention factor for muscle inflammatory response, through its effects on controlling intramuscular infiltration and phenotypes of monocytes/macrophages, on affecting accumulation and function of Treg cells, and on suppressing Th1 response in inflamed muscle. Our findings also imply an inhibition effect of estrogen on the intrinsic immune behaviors of muscle cells.
format Online
Article
Text
id pubmed-6661750
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-66617502019-08-05 Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells Liao, Zhao Hong Huang, Tao Xiao, Jiang Wei Gu, Rui Cai Ouyang, Jun Wu, Gang Liao, Hua Skelet Muscle Research BACKGROUND: Estrogen signaling is indispensable for muscle regeneration, yet the role of estrogen in the development of muscle inflammation, especially in the intramuscular T cell response, and the influence on the intrinsic immuno-behaviors of myofibers remain largely unknown. We investigated this issue using the mice model of cardiotoxin (CTX)-induced myoinjury, with or without estrogen level adjustment. METHODS: CTX injection i.m. (tibialis anterior, TA) was performed for preparing mice myoinjury model. Injection s.c. of 17β-estradiol (E(2)) or estrogen receptor antagonist 4-OHT, or ovariectomy (OVX), was used to change estrogen level of animal models in vivo. Serum E(2) level was evaluated by ELISA. Gene levels of estrogen receptor (ERs) and cytokines/chemokines in inflamed muscle were monitored by qPCR. Inflammatory infiltration was observed by immunofluorescence. Macrophage and T cell phenotypes were analyzed by FACS. Immunoblotting was used to assess protein levels of ERs and immunomolecules in C(2)C(12) myotubes treated with E(2) or 4-OHT, in the presence of IFN-γ. RESULTS: We monitored the increased serum E(2) level and the upregulated ERβ in regenerated myofibres after myotrauma. The absence of estrogen in vivo resulted in the more severe muscle inflammatory infiltration, involving the recruitment of monocyte/macrophage and CD4(+) T cells, and the heightened proinflammatory (M1) macrophage. Moreover, estrogen signaling loss led to Treg cells infiltration decrease, Th1 response elevation in inflamed muscle, and the markedly expression upregulation of immunomolecules in IFN-γ-stimulated C(2)C(12) myotubes in vitro. CONCLUSION: Our data suggest that estrogen is a positive intervention factor for muscle inflammatory response, through its effects on controlling intramuscular infiltration and phenotypes of monocytes/macrophages, on affecting accumulation and function of Treg cells, and on suppressing Th1 response in inflamed muscle. Our findings also imply an inhibition effect of estrogen on the intrinsic immune behaviors of muscle cells. BioMed Central 2019-07-29 /pmc/articles/PMC6661750/ /pubmed/31358063 http://dx.doi.org/10.1186/s13395-019-0205-2 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Liao, Zhao Hong
Huang, Tao
Xiao, Jiang Wei
Gu, Rui Cai
Ouyang, Jun
Wu, Gang
Liao, Hua
Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells
title Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells
title_full Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells
title_fullStr Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells
title_full_unstemmed Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells
title_short Estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and T cells
title_sort estrogen signaling effects on muscle-specific immune responses through controlling the recruitment and function of macrophages and t cells
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6661750/
https://www.ncbi.nlm.nih.gov/pubmed/31358063
http://dx.doi.org/10.1186/s13395-019-0205-2
work_keys_str_mv AT liaozhaohong estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells
AT huangtao estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells
AT xiaojiangwei estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells
AT guruicai estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells
AT ouyangjun estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells
AT wugang estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells
AT liaohua estrogensignalingeffectsonmusclespecificimmuneresponsesthroughcontrollingtherecruitmentandfunctionofmacrophagesandtcells