Skip to main content
Journal cover image

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

Publication ,  Journal Article
Luo, X; Chen, O; Wang, Z; Bang, S; Ji, J; Lee, SH; Huh, Y; Furutani, K; He, Q; Tao, X; Ko, M-C; Bortsov, A; Donnelly, CR; Chen, Y; Berta, T ...
Published in: Neuron
September 1, 2021

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 α (ERα) 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.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

Neuron

DOI

EISSN

1097-4199

Publication Date

September 1, 2021

Volume

109

Issue

17

Start / End Page

2691 / 2706.e5

Location

United States

Related Subject Headings

  • TRPV Cation Channels
  • Signal Transduction
  • Sex Factors
  • Nociceptors
  • Nociceptive Pain
  • Neurology & Neurosurgery
  • Nerve Fibers, Unmyelinated
  • Mice, Inbred C57BL
  • Mice
  • Male
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Luo, X., Chen, O., Wang, Z., Bang, S., Ji, J., Lee, S. H., … Ji, R.-R. (2021). IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice. Neuron, 109(17), 2691-2706.e5. https://doi.org/10.1016/j.neuron.2021.06.015
Luo, Xin, Ouyang Chen, Zilong Wang, Sangsu Bang, Jasmine Ji, Sang Hoon Lee, Yul Huh, et al. “IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice.Neuron 109, no. 17 (September 1, 2021): 2691-2706.e5. https://doi.org/10.1016/j.neuron.2021.06.015.
Luo X, Chen O, Wang Z, Bang S, Ji J, Lee SH, et al. IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice. Neuron. 2021 Sep 1;109(17):2691-2706.e5.
Luo, Xin, et al. “IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice.Neuron, vol. 109, no. 17, Sept. 2021, pp. 2691-2706.e5. Pubmed, doi:10.1016/j.neuron.2021.06.015.
Luo X, Chen O, Wang Z, Bang S, Ji J, Lee SH, Huh Y, Furutani K, He Q, Tao X, Ko M-C, Bortsov A, Donnelly CR, Chen Y, Nackley A, Berta T, Ji R-R. IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice. Neuron. 2021 Sep 1;109(17):2691-2706.e5.
Journal cover image

Published In

Neuron

DOI

EISSN

1097-4199

Publication Date

September 1, 2021

Volume

109

Issue

17

Start / End Page

2691 / 2706.e5

Location

United States

Related Subject Headings

  • TRPV Cation Channels
  • Signal Transduction
  • Sex Factors
  • Nociceptors
  • Nociceptive Pain
  • Neurology & Neurosurgery
  • Nerve Fibers, Unmyelinated
  • Mice, Inbred C57BL
  • Mice
  • Male