1,25-Dihydroxyvitamin D3 suppresses TLR8 expression and TLR8-mediated inflammatory responses in monocytes in vitro and experimental autoimmune encephalomyelitis in vivo

PLoS One. 2013;8(3):e58808. doi: 10.1371/journal.pone.0058808. Epub 2013 Mar 14.

Abstract

1,25-Dihydroxyvitamin D3 (1,25(OH)2D3) suppresses autoimmunity and inflammation; however, the mechanism of its action has not been fully understood. We sought in this study to determine whether the anti-immune/anti-inflammatory action of 1,25(OH)2D3 is in part mediated through an interplay between 1,25(OH)2D3 and toll-like receptor (TLR)7/8 signaling. 1,25(OH)2D3 treatment prior to and/or following experimental autoimmune encephalomyelitis (EAE) induction effectively reduced inflammatory cytokine expression in the spinal cord and ameliorated EAE. These effects were accompanied with a reduction in expression of several TLRs with the most profound effect observed for TLR8. The expression of TLR8 adaptor protein MyD88 was also significantly reduced by 1,25(OH)2D3. To determine the molecular mechanism by which 1,25(OH)2D3 suppresses EAE induction of TLR8 and inflammatory cytokine expression, we evaluated whether 1,25(OH)2D3 can directly inhibit TLR8 signaling and the resulting inflammatory responses in human THP-1 monocytes. 1,25(OH)2D3 treatment not only significantly reduced TLR8 expression but also the expression or activity of MyD88, IRF-4, IRF-7 and NF-kB in monocytes challenged with TLR8 ligands. TLR8 promoter-luciferase reporter assays indicated that 1,25(OH)2D3 decreases TLR8 mRNA level in part via inhibiting TLR8 gene transcription activity. As a result of inhibition on TLR8 signaling cascade at various stages, 1,25(OH)2D3 significantly diminished the TLR8 target gene expression (TNF-α and IL-1β). In summary, our novel findings suggest that TLR8 is a new target of 1,25(OH)2D3 and may mediate the anti-inflammatory action of 1,25(OH)2D3. Our findings also point to a destructive role of TLR8 in EAE and shed lights on pathogenesis of multiple sclerosis.

Publication types

  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Anti-Inflammatory Agents / pharmacology
  • Anti-Inflammatory Agents / therapeutic use
  • Calcitriol / pharmacology*
  • Calcitriol / therapeutic use
  • Cell Line
  • Down-Regulation / drug effects*
  • Encephalomyelitis, Autoimmune, Experimental / drug therapy
  • Encephalomyelitis, Autoimmune, Experimental / genetics*
  • Encephalomyelitis, Autoimmune, Experimental / metabolism*
  • Encephalomyelitis, Autoimmune, Experimental / pathology
  • Female
  • Humans
  • Inflammation / genetics
  • Inflammation / metabolism
  • Inflammation / pathology
  • Interleukin-1beta / metabolism
  • Ligands
  • Mice
  • Mice, Inbred C57BL
  • Monocytes / cytology
  • Monocytes / drug effects*
  • Monocytes / metabolism
  • Signal Transduction / drug effects
  • Spinal Cord / drug effects
  • Spinal Cord / metabolism
  • Toll-Like Receptor 8 / genetics*
  • Toll-Like Receptor 8 / metabolism*
  • Tumor Necrosis Factor-alpha / metabolism
  • Up-Regulation / drug effects

Substances

  • Anti-Inflammatory Agents
  • Interleukin-1beta
  • Ligands
  • TLR8 protein, mouse
  • Toll-Like Receptor 8
  • Tumor Necrosis Factor-alpha
  • Calcitriol

Grants and funding

This study was supported by funding from the Telemedicine and Advanced Technology Research Center (TATRC) at the US Army Medical Research and Materiel Command (USAMRMC) under Grant No. W81XWH-11-2-0052 (to David J. Baylink), GI Foundation of Loma Linda University (to Michael Walter) and Department of Medicine, Loma Linda University School of Medicine, (to Xuezhong Qin). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.