Abstract

Oxidized low density lipoprotein (oxLDL) induces macrophage activation, an event essential for atherosclerosis. Emerging evidence supports that epigenetic regulation plays important roles in macrophage activation and function. However, it remains unclear which epigenetic modulator is responsible for oxLDL-induced macrophage activation. Here, we identify for the first time KDM4A (JMJD2A) as an epigenetic modifying enzyme that controls oxLDL-induced pro-inflammatory M1 polarization of macrophages. OxLDL triggered M1 polarization of murine and human macrophages, characterized by expression of iNOS and robust production of inflammatory cytokines (e.g., TNF-α, MCP-1, IL-1β). In contrast, protein level of the M2 marker Arg1 was clearly decreased after treated with oxLDL. Notably, exposure to oxLDL resulted in markedly increased expression of KDM4A in macrophages. Functionally, shRNA knockdown of KDM4A significantly impaired M1 polarization and expression of inflammatory cytokines induced by oxLDL, accompanied by increased expression of Arg1 and VEGF. However, inhibition of KDM4A by shRNA or the pan-selective KDM inhibitor JIB-04 did not affect oxLDL-mediated activation of the NF-κB and hypoxia inducible factor (HIF) pathways, and vice versa. In addition, JIB-04 induced apoptosis of macrophages in a dose-dependent manner, an event attenuated by oxLDL. Together, these findings argue that KDM4A might represent a novel epigenetic modulator that acts to direct oxLDL-induced M1 polarization of macrophages, while its up-regulation is independent of NF-κB and HIF activation, two signals critical for pro-inflammatory activation of macrophages. They also suggest that KDM4A might serve as a potential target for epigenetic therapy in prevention and treatment of inflammatory diseases such as atherosclerosis.

Highlights

  • Atherosclerosis is recognized as a chronic inflammatory disease of the arterial wall, due to imbalance in lipid metabolism and uncontrolled inflammation [1]

  • A growing body of research has unveiled the roles of epigenetic regulation in a multitude of macrophage functions in a variety of inflammatory diseases, including atherosclerosis that has a major impact on human health

  • It remains unclear which histone epigenetic modifying enzyme is responsible for reprogramming M1 polarization of macrophages induced by Oxidized low density lipoprotein (oxLDL), an event essential for the pathogenesis of atherosclerosis www.impactjournals.com/oncotarget

Read more

Summary

Introduction

Atherosclerosis is recognized as a chronic inflammatory disease of the arterial wall, due to imbalance in lipid metabolism and uncontrolled inflammation [1]. The atherosclerotic process remains not fully understood, it has been well documented that macrophages play a central role throughout the pathogenesis of atherosclerosis [3] In this context, it is a general consensus that atherosclerosis is intrinsically a persistent inflammatory process primarily involving disproportion in macrophage polarization [1]. In response to environmental stimuli, macrophages polarize into either pro-inflammatory M1 or anti-inflammatory M2 phenotypes, in a context-specific manner The former is induced by multiple cytokines such as tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and granulocyte-macrophage colony-stimulating factor (GMCSF), which in turn produces a variety of cytokines (e.g., IL-6, IL-12, IL-1β, TNF-α, MCP-1, and IL-23) to promote inflammation [4]. M1 macrophages orchestrate the whole process of atherogenesis from formation of foam cells to plaque rupture, especially via evoking and sustaining local inflammatory response [3]

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