Abstract

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-κB and caspase 3-dependent cell death pathways. Loss- or gain-of-function experiments demonstrated that s-RNYs activate caspase 3 and NF-κB 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-κB-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.

Highlights

  • Macrophage apoptosis is a tightly regulated mechanism to control tissue homeostasis, resolution of inflammation[1,2] and innate immune response against microbial infection.[3]

  • In accordance with previous reports,[16,17] we found apoptosis and inflammation activation in monocytes/macrophages treated with pro-atherogenic stimuli, such as palmitic acid (PA) or oxidized LDL (oxLDL) in combination with thapsigargin (Tg) (Figure 1)

  • Small RNA derived from the RNY5 was not measured by stem-loop RT-qPCR because only the 3’ fragment is generated from this RNA in lipid-laden macrophages.[1]

Read more

Summary

Introduction

Macrophage apoptosis is a tightly regulated mechanism to control tissue homeostasis, resolution of inflammation[1,2] and innate immune response against microbial infection.[3]. By either inducing or inhibiting the expression, we have shown that s-RNYs activate both caspase-dependent cell death and NF-κB-dependent inflammation in cultured monocytes/macrophages. This phenotype was rescued by treating the monocytes/macrophages. The isolated s-RNY/Ro60 complex by affinity purification promoted TLR7-dependent apoptosis and inflammation when added in the medium of cultured monocytes/ macrophages. This effect was rescued by s-RNY antisense oligonucleotides and by TLR7 inhibitors. Overall, these data indicate that s-RNYs are intrinsic components of the machinery regulating lipid-laden macrophage phenotype and function as mediators of inflammation/apoptosis. Endothelial cells, which do not express TLR7 or s-RNYs when stimulated with pro-atherogenic agents,[1] are not activated by s-RNYs, confining the extracellular role of s-RNYs to immune cells

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call