Detailed Information

Cited 0 time in webofscience Cited 0 time in scopus
Metadata Downloads

RANKL-responsive epigenetic mechanism reprograms macrophages into bone-resorbing osteoclasts

Authors
Bae, SeyeonKim, KibyeongKang, KeunsooKim, HaeminLee, MinjoonOh, BrianKaneko, KaichiMa, SungkookChoi, Jae HoonKwak, HojoongLee, Eun YoungPark, Sung HoPark-Min, Kyung-Hyun
Issue Date
Jan-2023
Publisher
Springer Nature
Keywords
enhancer RNAs; Osteoclasts; Rheumatoid arthritis; super-enhancers
Citation
Cellular and Molecular Immunology, v.20, no.1, pp.94 - 109
Indexed
SCIE
SCOPUS
Journal Title
Cellular and Molecular Immunology
Volume
20
Number
1
Start Page
94
End Page
109
URI
https://scholarworks.bwise.kr/hanyang/handle/2021.sw.hanyang/182171
DOI
10.1038/s41423-022-00959-x
ISSN
1672-7681
Abstract
Monocyte/macrophage lineage cells are highly plastic and can differentiate into various cells under different environmental stimuli. Bone-resorbing osteoclasts are derived from the monocyte/macrophage lineage in response to receptor activator of NF-κB ligand (RANKL). However, the epigenetic signature contributing to the fate commitment of monocyte/macrophage lineage differentiation into human osteoclasts is largely unknown. In this study, we identified RANKL-responsive human osteoclast-specific superenhancers (SEs) and SE-associated enhancer RNAs (SE-eRNAs) by integrating data obtained from ChIP-seq, ATAC-seq, nuclear RNA-seq and PRO-seq analyses. RANKL induced the formation of 200 SEs, which are large clusters of enhancers, while suppressing 148 SEs in macrophages. RANKL-responsive SEs were strongly correlated with genes in the osteoclastogenic program and were selectively increased in human osteoclasts but marginally presented in osteoblasts, CD4+ T cells, and CD34+ cells. In addition to the major transcription factors identified in osteoclasts, we found that BATF binding motifs were highly enriched in RANKL-responsive SEs. The depletion of BATF1/3 inhibited RANKL-induced osteoclast differentiation. Furthermore, we found increased chromatin accessibility in SE regions, where RNA polymerase II was significantly recruited to induce the extragenic transcription of SE-eRNAs, in human osteoclasts. Knocking down SE-eRNAs in the vicinity of the NFATc1 gene diminished the expression of NFATc1, a major regulator of osteoclasts, and osteoclast differentiation. Inhibiting BET proteins suppressed the formation of some RANKL-responsive SEs and NFATc1-associated SEs, and the expression of SE-eRNA:NFATc1. Moreover, SE-eRNA:NFATc1 was highly expressed in the synovial macrophages of rheumatoid arthritis patients exhibiting high-osteoclastogenic potential. Our genome-wide analysis revealed RANKL-inducible SEs and SE-eRNAs as osteoclast-specific signatures, which may contribute to the development of osteoclast-specific therapeutic interventions.
Files in This Item
Go to Link
Appears in
Collections
서울 자연과학대학 > 서울 생명과학과 > 1. Journal Articles

qrcode

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

Related Researcher

Researcher Choi, Jae Hoon photo

Choi, Jae Hoon
COLLEGE OF NATURAL SCIENCES (DEPARTMENT OF LIFE SCIENCE)
Read more

Altmetrics

Total Views & Downloads

BROWSE