Cargando…

Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling

BACKGROUND: To explore the role of skeletal muscle specific TGF-β signaling on macrophages efferocytosis in inflamed muscle caused by Cardiotoxin (CTX) injection. METHODS: CTX myoinjury was manipulated in TGF-βr2(flox/flox) (control) mice or transgenic mice with TGF-β receptor 2 (TGF-βr2) being spec...

Descripción completa

Detalles Bibliográficos
Autores principales: Liao, Zhaohong, Lan, Haiqiang, Jian, Xiaoting, Huang, Jingwen, Wang, Han, Hu, Jijie, Liao, Hua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10318691/
https://www.ncbi.nlm.nih.gov/pubmed/37403092
http://dx.doi.org/10.1186/s12964-023-01163-8
_version_ 1785068091978285056
author Liao, Zhaohong
Lan, Haiqiang
Jian, Xiaoting
Huang, Jingwen
Wang, Han
Hu, Jijie
Liao, Hua
author_facet Liao, Zhaohong
Lan, Haiqiang
Jian, Xiaoting
Huang, Jingwen
Wang, Han
Hu, Jijie
Liao, Hua
author_sort Liao, Zhaohong
collection PubMed
description BACKGROUND: To explore the role of skeletal muscle specific TGF-β signaling on macrophages efferocytosis in inflamed muscle caused by Cardiotoxin (CTX) injection. METHODS: CTX myoinjury was manipulated in TGF-βr2(flox/flox) (control) mice or transgenic mice with TGF-β receptor 2 (TGF-βr2) being specifically deleted in skeletal muscle (SM TGF-βr2(−/−)). Gene levels of TGF-β signal molecules, special inflammatory mediators in damaged muscle or in cultured and differentiated myogenic precursor cells (MPC-myotubes) were monitored by transcriptome microarray or qRT-PCR. TGF-β pathway molecules, myokines and embryonic myosin heavy chain in regenerating myofibers, the phenotype and efferocytosis of macrophages were evaluated by immunofluorescence, immunoblotting, Luminex, or FACS analysis. In vitro apoptotic cells were prepared by UV-irradiation. RESULTS: In control mice, TGF-β-Smad2/3 signaling were significantly up-regulated in regenerating centronuclear myofibers after CTX-myoinjury. More severe muscle inflammation was caused by the deficiency of muscle TGF-β signaling, with the increased number of M1, but the decreased number of M2 macrophages. Notably, the deficiency of TGF-β signaling in myofibers dramatically affected on the ability of macrophages to conduct efferocytosis, marked by the decreased number of Annexin-V(−)F4/80(+)Tunel(+) macrophages in inflamed muscle, and the impaired uptake of macrophages to PKH67(+) apoptotic cells transferred into damaged muscle. Further, our study suggested that, the intrinsic TGF-β signaling directed IL-10-Vav1-Rac1 efferocytosis signaling in muscle macrophages. CONCLUSIONS: Our data demonstrate that muscle inflammation can be suppressed potentially by activating the intrinsic TGF-β signaling in myofibers to promote IL-10 dependent-macrophages efferocytosis. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12964-023-01163-8.
format Online
Article
Text
id pubmed-10318691
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103186912023-07-05 Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling Liao, Zhaohong Lan, Haiqiang Jian, Xiaoting Huang, Jingwen Wang, Han Hu, Jijie Liao, Hua Cell Commun Signal Research BACKGROUND: To explore the role of skeletal muscle specific TGF-β signaling on macrophages efferocytosis in inflamed muscle caused by Cardiotoxin (CTX) injection. METHODS: CTX myoinjury was manipulated in TGF-βr2(flox/flox) (control) mice or transgenic mice with TGF-β receptor 2 (TGF-βr2) being specifically deleted in skeletal muscle (SM TGF-βr2(−/−)). Gene levels of TGF-β signal molecules, special inflammatory mediators in damaged muscle or in cultured and differentiated myogenic precursor cells (MPC-myotubes) were monitored by transcriptome microarray or qRT-PCR. TGF-β pathway molecules, myokines and embryonic myosin heavy chain in regenerating myofibers, the phenotype and efferocytosis of macrophages were evaluated by immunofluorescence, immunoblotting, Luminex, or FACS analysis. In vitro apoptotic cells were prepared by UV-irradiation. RESULTS: In control mice, TGF-β-Smad2/3 signaling were significantly up-regulated in regenerating centronuclear myofibers after CTX-myoinjury. More severe muscle inflammation was caused by the deficiency of muscle TGF-β signaling, with the increased number of M1, but the decreased number of M2 macrophages. Notably, the deficiency of TGF-β signaling in myofibers dramatically affected on the ability of macrophages to conduct efferocytosis, marked by the decreased number of Annexin-V(−)F4/80(+)Tunel(+) macrophages in inflamed muscle, and the impaired uptake of macrophages to PKH67(+) apoptotic cells transferred into damaged muscle. Further, our study suggested that, the intrinsic TGF-β signaling directed IL-10-Vav1-Rac1 efferocytosis signaling in muscle macrophages. CONCLUSIONS: Our data demonstrate that muscle inflammation can be suppressed potentially by activating the intrinsic TGF-β signaling in myofibers to promote IL-10 dependent-macrophages efferocytosis. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12964-023-01163-8. BioMed Central 2023-07-04 /pmc/articles/PMC10318691/ /pubmed/37403092 http://dx.doi.org/10.1186/s12964-023-01163-8 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
Liao, Zhaohong
Lan, Haiqiang
Jian, Xiaoting
Huang, Jingwen
Wang, Han
Hu, Jijie
Liao, Hua
Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling
title Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling
title_full Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling
title_fullStr Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling
title_full_unstemmed Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling
title_short Myofiber directs macrophages IL-10-Vav1-Rac1 efferocytosis pathway in inflamed muscle following CTX myoinjury by activating the intrinsic TGF-β signaling
title_sort myofiber directs macrophages il-10-vav1-rac1 efferocytosis pathway in inflamed muscle following ctx myoinjury by activating the intrinsic tgf-β signaling
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10318691/
https://www.ncbi.nlm.nih.gov/pubmed/37403092
http://dx.doi.org/10.1186/s12964-023-01163-8
work_keys_str_mv AT liaozhaohong myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling
AT lanhaiqiang myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling
AT jianxiaoting myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling
AT huangjingwen myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling
AT wanghan myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling
AT hujijie myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling
AT liaohua myofiberdirectsmacrophagesil10vav1rac1efferocytosispathwayininflamedmusclefollowingctxmyoinjurybyactivatingtheintrinsictgfbsignaling