Abstract

This study was designed to examine the role of hydrogen sulfide (H2S) in the generation of oxidized low-density lipoprotein (ox-LDL)-stimulated monocyte chemoattractant protein 1 (MCP-1) from macrophages and possible mechanisms. THP-1 cells and RAW macrophages were pretreated with sodium hydrosulfide (NaHS) and hexyl acrylate and then treated with ox-LDL. The results showed that ox-LDL treatment down-regulated the H2S/cystathionine-β-synthase pathway, with increased MCP-1 protein and mRNA expression in both THP-1 cells and RAW macrophages. Hexyl acrylate promoted ox-LDL-induced inflammation, whereas the H2S donor NaHS inhibited it. NaHS markedly suppressed NF-κB p65 phosphorylation, nuclear translocation, DNA binding activity, and recruitment to the MCP-1 promoter in ox-LDL-treated macrophages. Furthermore, NaHS decreased the ratio of free thiol groups in p65, whereas the thiol reductant DTT reversed the inhibiting effect of H2S on the p65 DNA binding activity. Most importantly, site-specific mutation of cysteine 38 to serine in p65 abolished the effect of H2S on the sulfhydration of NF-κB and ox-LDL-induced NF-κB activation. These results suggested that endogenous H2S inhibited ox-LDL-induced macrophage inflammation by suppressing NF-κB p65 phosphorylation, nuclear translocation, DNA binding activity, and recruitment to the MCP-1 promoter. The sulfhydration of free thiol group on cysteine 38 in p65 served as a molecular mechanism by which H2S inhibited NF-κB pathway activation in ox-LDL-induced macrophage inflammation.

Highlights

  • IntroductionResults: H2S inhibits NF-␬B p65 phosphorylation, nuclear translocation, DNA binding activity, and recruitment to monocyte chemoattractant protein 1 (MCP-1) promoter in oxidized low-density lipoprotein (ox-LDL)-treated macrophages by targeting the free sulfhydryl group on cysteine 38 in p65

  • The mechanisms by which H2S regulates inflammation remain unclear

  • Changes in Endogenous H2S Pathway in Ox-LDL-induced, THP-1-derived Macrophages—We detected the changes in the H2S level and H2S synthase expression depending on the duration of treatment with oxidized low-density lipoprotein (ox-LDL)

Read more

Summary

Introduction

Results: H2S inhibits NF-␬B p65 phosphorylation, nuclear translocation, DNA binding activity, and recruitment to MCP-1 promoter in ox-LDL-treated macrophages by targeting the free sulfhydryl group on cysteine 38 in p65. NaHS markedly suppressed NF-␬B p65 phosphorylation, nuclear translocation, DNA binding activity, and recruitment to the MCP-1 promoter in ox-LDL-treated macrophages. Site-specific mutation of cysteine 38 to serine in p65 abolished the effect of H2S on the sulfhydration of NF-␬B and ox-LDL-induced NF-␬B activation These results suggested that endogenous H2S inhibited ox-LDL-induced macrophage inflammation by suppressing NF-␬B p65 phosphorylation, nuclear translocation, DNA binding activity, and recruitment to the MCP-1 promoter. The sulfhydration of free thiol group on cysteine 38 in p65 served as a molecular mechanism by which H2S inhibited NF-␬B pathway activation in ox-LDL-induced macrophage inflammation

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