4.8 Article

IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice

Journal

NEURON
Volume 109, Issue 17, Pages 2691-+

Publisher

CELL PRESS
DOI: 10.1016/j.neuron.2021.06.015

Keywords

-

Categories

Funding

  1. Duke University Anesthesiology Research Funds
  2. NIH [AR069861, DE027454, NS113243]
  3. [T32 GM008600]

Ask authors/readers for more resources

The IL-23/IL-17A/TRPV1 axis regulates female-specific mechanical pain, with IL-23 inducing pain in females but impaired in mice lacking IL-23 or IL-23r. IL-23-induced pain is promoted by estrogen and suppressed by androgen.
Although sex dimorphism is increasingly recognized as an important factor in pain, female-specific pain signaling is not well studied. Here we report that administration of IL-23 produces mechanical pain (mechanical allodynia) in female but not male mice, and chemotherapy-induced mechanical pain is selectively impaired in female mice lacking Il23 or Il23r. IL-23-induced pain is promoted by estrogen but suppressed by androgen, suggesting an involvement of sex hormones. IL-23 requires C-fiber nociceptors and TRPV1 to produce pain but does not directly activate nociceptor neurons. Notably, IL-23 requires IL-17A release from macrophages to evoke mechanical pain in females. Low-dose IL-17A directly activates nociceptors and induces mechanical pain only in females. Finally, deletion of estrogen receptor subunit a (ERa) in TRPV1(+) nociceptors abolishes IL -23-and IL-17-induced pain in females. These findings demonstrate that the IL-23/IL-17A/TRPV1 axis regulates female-specific mechanical pain via neuro-immune interactions. Our study also reveals sex dimorphism at both immune and neuronal levels.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available