Ferryl Hemoglobin Inhibits Osteoclastic Differentiation of Macrophages in Hemorrhaged Atherosclerotic Plaques

Oxid Med Cell Longev. 2020 Feb 27:2020:3721383. doi: 10.1155/2020/3721383. eCollection 2020.

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.

MeSH terms

  • Animals
  • Bone Resorption / pathology
  • Calcinosis
  • Carotid Arteries / drug effects
  • Carotid Arteries / pathology
  • Cell Differentiation* / drug effects
  • Down-Regulation / drug effects
  • Gene Expression Regulation / drug effects
  • Heme Oxygenase-1 / metabolism
  • Hemoglobins / pharmacology*
  • Hemorrhage / pathology*
  • Humans
  • Macrophages / drug effects
  • Macrophages / metabolism
  • Macrophages / pathology*
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Models, Biological
  • Osteoclasts / drug effects
  • Osteoclasts / metabolism
  • Osteoclasts / pathology*
  • Osteogenesis / drug effects
  • Oxidation-Reduction / drug effects
  • Plaque, Atherosclerotic / genetics
  • Plaque, Atherosclerotic / pathology*
  • Protein Binding / drug effects
  • RANK Ligand / genetics
  • RANK Ligand / metabolism
  • RAW 264.7 Cells
  • Receptor Activator of Nuclear Factor-kappa B / metabolism
  • Signal Transduction / drug effects

Substances

  • Hemoglobins
  • RANK Ligand
  • Receptor Activator of Nuclear Factor-kappa B
  • TNFRSF11A protein, human
  • ferrylhemoglobin
  • Heme Oxygenase-1