Article (Scientific journals)
RNY (YRNA)-derived small RNAs regulate cell death and inflammation in monocytes/macrophages.
Hizir, Zoheir; Bottini, Silvia; Grandjean, Valerie et al.
2017In Cell Death and Disease, 8 (1), p. 2530
Peer Reviewed verified by ORBi
 

Files


Full Text
Hizir et al 2017.pdf
Publisher postprint (1.36 MB)
Download

All documents in ORBi are protected by a user license.

Send to



Details



Keywords :
Animals; Apoptosis/genetics; Atherosclerosis/genetics/metabolism/pathology; Autoantigens/genetics/metabolism; Caspase 3/genetics/metabolism; Humans; Inflammation/genetics/metabolism/pathology; Macrophages/metabolism/pathology; Mice; Monocytes/metabolism/pathology; RNA, Small Cytoplasmic/genetics/metabolism; Ribonucleoproteins/genetics/metabolism; Toll-Like Receptor 7/genetics
Abstract :
[en] The recent discovery of new classes of small RNAs has opened unknown territories to explore new regulations of physiopathological events. We have recently demonstrated that RNY (or Y RNA)-derived small RNAs (referred to as s-RNYs) are an independent class of clinical biomarkers to detect coronary artery lesions and are associated with atherosclerosis burden. Here, we have studied the role of s-RNYs in human and mouse monocytes/macrophages and have shown that in lipid-laden monocytes/macrophages s-RNY expression is timely correlated to the activation of both NF-kappaB and caspase 3-dependent cell death pathways. Loss- or gain-of-function experiments demonstrated that s-RNYs activate caspase 3 and NF-kappaB signaling pathways ultimately promoting cell death and inflammatory responses. As, in atherosclerosis, Ro60-associated s-RNYs generated by apoptotic macrophages are released in the blood of patients, we have investigated the extracellular function of the s-RNY/Ro60 complex. Our data demonstrated that s-RNY/Ro60 complex induces caspase 3-dependent cell death and NF-kappaB-dependent inflammation, when added to the medium of cultured monocytes/macrophages. Finally, we have shown that s-RNY function is mediated by Toll-like receptor 7 (TLR7). Indeed using chloroquine, which disrupts signaling of endosome-localized TLRs 3, 7, 8 and 9 or the more specific TLR7/9 antagonist, the phosphorothioated oligonucleotide IRS954, we blocked the effect of either intracellular or extracellular s-RNYs. These results position s-RNYs as relevant novel functional molecules that impacts on macrophage physiopathology, indicating their potential role as mediators of inflammatory diseases, such as atherosclerosis.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
Hizir, Zoheir ;  Université de Liège - ULiège > Département des sciences biomédicales et précliniques > GIGA-R : Labo of Molecular Immunol. and Signal Transduction
Bottini, Silvia
Grandjean, Valerie
Trabucchi, Michele
Repetto, Emanuela
Language :
English
Title :
RNY (YRNA)-derived small RNAs regulate cell death and inflammation in monocytes/macrophages.
Publication date :
2017
Journal title :
Cell Death and Disease
eISSN :
2041-4889
Publisher :
Nature Publishing Group, London, United Kingdom
Volume :
8
Issue :
1
Pages :
e2530
Peer reviewed :
Peer Reviewed verified by ORBi
Available on ORBi :
since 04 June 2018

Statistics


Number of views
72 (0 by ULiège)
Number of downloads
66 (0 by ULiège)

Scopus citations®
 
51
Scopus citations®
without self-citations
48
OpenCitations
 
42

Bibliography


Similar publications



Contact ORBi