4.6 Article

Sex-dependent role of microglia in disulfide high mobility group box 1 protein-mediated mechanical hypersensitivity

期刊

PAIN
卷 162, 期 2, 页码 446-458

出版社

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1097/j.pain.0000000000002033

关键词

HMGB1; Microglia; Sex dimorphism; Spinal; LC-MS; MS; Minocycline; Pain

资金

  1. Swedish Research Council [20138373, 2015-02776]
  2. Ragnar Soderberg Foundation
  3. Knut and Alice Wallenberg Foundation
  4. William K. Bowes Foundation
  5. European Union [602919]
  6. European Union Horizon 2020 research and innovation program under the Marie Sklodowska-Curie grant [642720]
  7. ALF grants Region Uppsala
  8. Magnus Bergvall Foundation
  9. Family Lundblad Foundation
  10. NIH [NS098826, NS096030]
  11. Strategic Funds Uppsala University Hospital
  12. Swedish Research Council [2015-02776] Funding Source: Swedish Research Council

向作者/读者索取更多资源

The study explored the differential effects of TLR4-activating HMGB1 on microglia in male and female mice, finding higher cytokine and chemokine expression in males. Only male mice were protected from HMGB1-induced mechanical hypersensitivity with TLR4 ablation in myeloid-derived cells and minocycline treatment. Liquid chromatography-mass spectrometry revealed upregulation of antinociceptive proteins in male mice after minocycline administration, with alpha-1-antitrypsin offering partial protection against HMGB1-induced pain specifically in males.
High mobility group box 1 protein (HMGB1) is increasingly regarded as an important player in the spinal regulation of chronic pain. Although it has been reported that HMGB1 induces spinal glial activation in a Toll-like receptor (TLR)4-dependent fashion, the aspect of sexual dimorphisms has not been thoroughly addressed. Here, we examined whether the action of TLR4-activating, partially reduced disulfide HMGB1 on microglia induces nociceptive behaviors in a sex-dependent manner. We found disulfide HMGB1 to equally increase microglial Iba1 immunoreactivity in lumbar spinal dorsal horn in male and female mice, but evoke higher cytokine and chemokine expression in primary microglial culture derived from males compared to females. Interestingly, TLR4 ablation in myeloid-derived cells, which include microglia, only protected male mice from developing HMGB1-induced mechanical hypersensitivity. Spinal administration of the glial inhibitor, minocycline, with disulfide HMGB1 also prevented pain-like behavior in male mice. To further explore sex difference, we examined the global spinal protein expression using liquid chromatography-mass spectrometry and found several antinociceptive and anti-inflammatory proteins to be upregulated in only male mice subjected to minocycline. One of the proteins elevated, alpha-1-antitrypsin, partially protected males but not females from developing HMGB1-induced pain. Targeting downstream proteins of alpha-1-antitrypsin failed to produce robust sex differences in pain-like behavior, suggesting that several proteins identified by liquid chromatography-mass spectrometry are required to modulate the effects. Taken together, the current study highlights the importance of mapping sex dimorphisms in pain mechanisms and point to processes potentially involved in the spinal antinociceptive effect of microglial inhibition in male mice.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据