Cargando…

RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling

BACKGROUND: The cytoskeletal architecture of osteoclasts (OCs) and bone resorption activity must be appropriately controlled for proper bone remodeling, which is associated with osteoporosis. The RhoA protein of GTPase plays a regulatory role in cytoskeletal components and contributes to osteoclast...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Jirong, Xu, Chengyun, Zhang, Jing, Bao, Yizhong, Tang, Ying, Lv, Xiaoling, Ma, Bo, Wu, Ximei, Mao, Genxiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10077675/
https://www.ncbi.nlm.nih.gov/pubmed/37020186
http://dx.doi.org/10.1186/s10020-023-00638-1
_version_ 1785020355149037568
author Wang, Jirong
Xu, Chengyun
Zhang, Jing
Bao, Yizhong
Tang, Ying
Lv, Xiaoling
Ma, Bo
Wu, Ximei
Mao, Genxiang
author_facet Wang, Jirong
Xu, Chengyun
Zhang, Jing
Bao, Yizhong
Tang, Ying
Lv, Xiaoling
Ma, Bo
Wu, Ximei
Mao, Genxiang
author_sort Wang, Jirong
collection PubMed
description BACKGROUND: The cytoskeletal architecture of osteoclasts (OCs) and bone resorption activity must be appropriately controlled for proper bone remodeling, which is associated with osteoporosis. The RhoA protein of GTPase plays a regulatory role in cytoskeletal components and contributes to osteoclast adhesion, podosome positioning, and differentiation. Although osteoclast investigations have traditionally been performed by in vitro analysis, however, the results have been inconsistent, and the significance of RhoA in bone physiology and pathology is still unknown. METHODS: We generated RhoA knockout mice by specifically deleting RhoA in the osteoclast lineage to understand more about RhoA’s involvement in bone remodeling. The function of RhoA in osteoclast differentiation and bone resorption and the mechanisms were assessed using bone marrow macrophages (BMMs) in vitro. The ovariectomized (OVX) mouse model was adopted to examine the pathological effect of RhoA in bone loss. RESULTS: Conditional deletion of RhoA in the osteoclast lineage causes a severe osteopetrosis phenotype, which is attributable to a bone resorption suppression. Further mechanistic studies suggest that RhoA deficiency suppresses Akt-mTOR-NFATc1 signaling during osteoclast differentiation. Additionally, RhoA activation is consistently related to the significant enhancement the osteoclast activity, which culminates in the development of an osteoporotic bone phenotype. Furthermore, in mice, the absence of RhoA in osteoclast precursors prevented occurring OVX-induced bone loss. CONCLUSION: RhoA promoted osteoclast development via the Akt-mTOR-NFATc1 signaling pathway, resulting a osteoporosis phenotype, and that manipulating RhoA activity might be a therapeutic strategy for osteoporotic bone loss. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s10020-023-00638-1.
format Online
Article
Text
id pubmed-10077675
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100776752023-04-07 RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling Wang, Jirong Xu, Chengyun Zhang, Jing Bao, Yizhong Tang, Ying Lv, Xiaoling Ma, Bo Wu, Ximei Mao, Genxiang Mol Med Research Article BACKGROUND: The cytoskeletal architecture of osteoclasts (OCs) and bone resorption activity must be appropriately controlled for proper bone remodeling, which is associated with osteoporosis. The RhoA protein of GTPase plays a regulatory role in cytoskeletal components and contributes to osteoclast adhesion, podosome positioning, and differentiation. Although osteoclast investigations have traditionally been performed by in vitro analysis, however, the results have been inconsistent, and the significance of RhoA in bone physiology and pathology is still unknown. METHODS: We generated RhoA knockout mice by specifically deleting RhoA in the osteoclast lineage to understand more about RhoA’s involvement in bone remodeling. The function of RhoA in osteoclast differentiation and bone resorption and the mechanisms were assessed using bone marrow macrophages (BMMs) in vitro. The ovariectomized (OVX) mouse model was adopted to examine the pathological effect of RhoA in bone loss. RESULTS: Conditional deletion of RhoA in the osteoclast lineage causes a severe osteopetrosis phenotype, which is attributable to a bone resorption suppression. Further mechanistic studies suggest that RhoA deficiency suppresses Akt-mTOR-NFATc1 signaling during osteoclast differentiation. Additionally, RhoA activation is consistently related to the significant enhancement the osteoclast activity, which culminates in the development of an osteoporotic bone phenotype. Furthermore, in mice, the absence of RhoA in osteoclast precursors prevented occurring OVX-induced bone loss. CONCLUSION: RhoA promoted osteoclast development via the Akt-mTOR-NFATc1 signaling pathway, resulting a osteoporosis phenotype, and that manipulating RhoA activity might be a therapeutic strategy for osteoporotic bone loss. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s10020-023-00638-1. BioMed Central 2023-04-05 /pmc/articles/PMC10077675/ /pubmed/37020186 http://dx.doi.org/10.1186/s10020-023-00638-1 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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/) .
spellingShingle Research Article
Wang, Jirong
Xu, Chengyun
Zhang, Jing
Bao, Yizhong
Tang, Ying
Lv, Xiaoling
Ma, Bo
Wu, Ximei
Mao, Genxiang
RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling
title RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling
title_full RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling
title_fullStr RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling
title_full_unstemmed RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling
title_short RhoA promotes osteoclastogenesis and regulates bone remodeling through mTOR-NFATc1 signaling
title_sort rhoa promotes osteoclastogenesis and regulates bone remodeling through mtor-nfatc1 signaling
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10077675/
https://www.ncbi.nlm.nih.gov/pubmed/37020186
http://dx.doi.org/10.1186/s10020-023-00638-1
work_keys_str_mv AT wangjirong rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT xuchengyun rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT zhangjing rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT baoyizhong rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT tangying rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT lvxiaoling rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT mabo rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT wuximei rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling
AT maogenxiang rhoapromotesosteoclastogenesisandregulatesboneremodelingthroughmtornfatc1signaling