[HTML][HTML] Heme molecule functions as an endogenous agonist of astrocyte TLR2 to contribute to secondary brain damage after intracerebral hemorrhage

H Min, B Choi, YH Jang, IH Cho, SJ Lee - Molecular brain, 2017 - Springer
H Min, B Choi, YH Jang, IH Cho, SJ Lee
Molecular brain, 2017Springer
Abstract Toll-like receptor 2 (TLR2) was recently shown to contribute to secondary brain
damage after intracerebral hemorrhage (ICH), although the molecular mechanisms of this
contribution are elusive. In this study, we tested the hypothesis that hemin functions as a
TLR2 endogenous agonist, causing proinflammatory astrocyte activation and secondary
brain damage after ICH. Hemin administration to the mouse brain striatum induced ICH
injury and neurological deficits, however, the brain injury volume and neurological deficits …
Abstract
Toll-like receptor 2 (TLR2) was recently shown to contribute to secondary brain damage after intracerebral hemorrhage (ICH), although the molecular mechanisms of this contribution are elusive. In this study, we tested the hypothesis that hemin functions as a TLR2 endogenous agonist, causing proinflammatory astrocyte activation and secondary brain damage after ICH. Hemin administration to the mouse brain striatum induced ICH injury and neurological deficits, however, the brain injury volume and neurological deficits due to hemin injection were significantly reduced in TLR2 knock-out (KO) mice. Hemin administration induced neutrophil infiltration and upregulated neutrophil-attracting chemokine and proinflammatory cytokine expression in wild-type (WT) mice; these effects were ameliorated in TLR2 KO mice. Likewise, ICH-induced blood-brain barrier (BBB) damage was also decreased in TLR2 KO mice. This effect was most likely due to reduced matrix metalloproteinase 9 (MMP9) activity in the TLR2 KO mice compared to WT mice. In primary astrocytes, hemin directly induced MMP9 activity as well as proinflammatory cytokine and chemokine expression in a TLR2-dependent manner. Finally, hemin-induced MMP9 activity and proinflammatory gene expression were almost completely blocked by TLR2-neutralizing antibodies. Taken together, our data propose that heme released to the brain parenchyma after ICH injury activates TLR2 in astrocytes and induces inflammatory gene expression and BBB damage, which contribute to secondary brain damage after ICH.
Springer