Abstract

Intraplaque hemorrhage frequently occurs in atherosclerotic plaques resulting in cell-free hemoglobin, which is oxidized to ferryl hemoglobin (FHb) in the highly oxidative environment. Osteoclast-like cells (OLCs) derived from macrophages signify a counterbalance mechanism for calcium deposition in atherosclerosis. Our aim was to investigate whether oxidized hemoglobin alters osteoclast formation, thereby affecting calcium removal from mineralized atherosclerotic lesions. RANKL- (receptor activator of nuclear factor kappa-Β ligand-) induced osteoclastogenic differentiation and osteoclast activity of RAW264.7 cells were studied in response to oxidized hemoglobin via assessing bone resorption activity, expression of osteoclast-specific genes, and the activation of signalization pathways. OLCs in diseased human carotid arteries were assessed by immunohistochemistry. FHb, but not ferrohemoglobin, decreased bone resorption activity and inhibited osteoclast-specific gene expression (tartrate-resistant acid phosphatase, calcitonin receptor, and dendritic cell-specific transmembrane protein) induced by RANKL. In addition, FHb inhibited osteoclastogenic signaling pathways downstream of RANK (receptor activator of nuclear factor kappa-Β). It prevented the induction of TRAF6 (tumor necrosis factor (TNF) receptor-associated factor 6) and c-Fos, phosphorylation of p-38 and JNK (c-Jun N-terminal kinase), and nuclear translocation of NFκB (nuclear factor kappa-Β) and NFATc1 (nuclear factor of activated T-cells, cytoplasmic 1). These effects were independent of heme oxygenase-1 demonstrated by knocking down HO-1 gene in RAW264.7 cells and in mice. Importantly, FHb competed with RANK for RANKL binding suggesting possible mechanisms by which FHb impairs osteoclastic differentiation. In diseased human carotid arteries, OLCs were abundantly present in calcified plaques and colocalized with regions of calcium deposition, while the number of these cells were lower in hemorrhagic lesions exhibiting accumulation of FHb despite calcium deposition. We conclude that FHb inhibits RANKL-induced osteoclastic differentiation of macrophages and suggest that accumulation of FHb in a calcified area of atherosclerotic lesion with hemorrhage retards the formation of OLCs potentially impairing calcium resorption.

Highlights

  • Cardiovascular disease is the leading cause of death worldwide [1] and vascular calcification is one of the independent risk factors associated with such morbidity and mortality [2,3,4,5]

  • Induced osteoclastogenesis, murine macrophage RAW264.7 cells were cultured in osteoclastogenic medium containing 50 ng/mL receptor activator of nuclear factor kappa-Β ligand (RANKL) in the presence or absence of Hb, MetHb, or ferryl hemoglobin (FHb)

  • We analyzed the effect of the different Hb forms on RANKL-induced osteoclastogenesis using tartrate-resistant acid phosphatase (TRAP) staining (Figure 1(a))

Read more

Summary

Introduction

Cardiovascular disease is the leading cause of death worldwide [1] and vascular calcification is one of the independent risk factors associated with such morbidity and mortality [2,3,4,5]. Pathogenesis of vascular calcification is an active, finely tuned process with many similarities to the mechanism of skeletal bone formation [6]. Oxidative Medicine and Cellular Longevity by osteoblasts and bone resorption by osteoclasts (OCs) are synchronized processes [7]. OCs are multinucleated cells derived from monocyte/macrophage lineage specialized for bone resorption [10]. The differentiation of macrophages to OCs requires macrophage/monocyte colony-stimulating factor-1 (M-CSF) and the receptor activator of nuclear factor kappa-Β ligand (RANKL) [11]. RANKL interacts with the receptor activator of nuclear factor kappa-B (RANK) activating downstream genes including tumor necrosis factor (TNF) receptorassociated factor 6 (TRAF6) [12], c-Jun N-terminal kinase (JNK) [13], p38 [14], nuclear factor-kappa B (NFκB) [15], c-Fos [16], and nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) [17]. NFATc1 plays a pivotal role in OC maturation by regulating the expression of OCspecific genes, such as dendritic cell-specific transmembrane protein (DC-STAMP), tartrate-resistant acid phosphatase (TRAP), cathepsin K (CatK), and calcitonin receptor (CTR) [18,19,20,21,22]

Objectives
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