18 214

Cited 36 times in

Selenoprotein W ensures physiological bone remodeling by preventing hyperactivity of osteoclasts

Authors
 Hyunsoo Kim  ;  Kyunghee Lee  ;  Jin Man Kim  ;  Mi Yeong Kim  ;  Jae-Ryong Kim  ;  Han-Woong Lee  ;  Youn Wook Chung  ;  Hong-In Shin  ;  Taesoo Kim  ;  Eui-Soon Park  ;  Jaerang Rho  ;  Seoung Hoon Lee  ;  Nacksung Kim  ;  Soo Young Lee  ;  Yongwon Choi  ;  Daewon Jeong 
Citation
 NATURE COMMUNICATIONS, Vol.12(1) : 2258, 2021-04 
Journal Title
NATURE COMMUNICATIONS
Issue Date
2021-04
MeSH
14-3-3 Proteins / metabolism ; Animals ; Bone Remodeling / genetics* ; Cell Differentiation / genetics ; Disease Models, Animal ; Gene Expression Regulation / physiology ; Humans ; Male ; Mice ; Mice, Knockout ; NFATC Transcription Factors / metabolism ; Osteoclasts / physiology* ; Osteogenesis / genetics* ; Osteoporosis / genetics* ; Osteoporosis / pathology ; RANK Ligand / metabolism ; RNA-Seq ; Selenoprotein W / genetics ; Selenoprotein W / metabolism* ; Signal Transduction / physiology ; TNF Receptor-Associated Factor 6 / metabolism
Abstract
Selenoproteins containing selenium in the form of selenocysteine are critical for bone remodeling. However, their underlying mechanism of action is not fully understood. Herein, we report the identification of selenoprotein W (SELENOW) through large-scale mRNA profiling of receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation, as a protein that is downregulated via RANKL/RANK/tumour necrosis factor receptor-associated factor 6/p38 signaling. RNA-sequencing analysis revealed that SELENOW regulates osteoclastogenic genes. SELENOW overexpression enhances osteoclastogenesis in vitro via nuclear translocation of NF-κB and nuclear factor of activated T-cells cytoplasmic 1 mediated by 14-3-3γ, whereas its deficiency suppresses osteoclast formation. SELENOW-deficient and SELENOW-overexpressing mice exhibit high bone mass phenotype and osteoporosis, respectively. Ectopic SELENOW expression stimulates cell-cell fusion critical for osteoclast maturation as well as bone resorption. Thus, RANKL-dependent repression of SELENOW regulates osteoclast differentiation and blocks osteoporosis caused by overactive osteoclasts. These findings demonstrate a biological link between selenium and bone metabolism.
Files in This Item:
T202125558.pdf Download
DOI
10.1038/s41467-021-22565-7
Appears in Collections:
1. College of Medicine (의과대학) > BioMedical Science Institute (의생명과학부) > 1. Journal Papers
Yonsei Authors
Chung, Youn Wook(정연욱) ORCID logo https://orcid.org/0000-0002-4382-1410
URI
https://ir.ymlib.yonsei.ac.kr/handle/22282913/190985
사서에게 알리기
  feedback

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

Browse

Links