Skip to main content
construction release_alert
Scholars@Duke will be undergoing maintenance April 11-15. Some features may be unavailable during this time.
cancel

IL-23 Enhances C-Fiber-Mediated and Blue Light-Induced Spontaneous Pain in Female Mice.

Publication ,  Journal Article
Ji, J; He, Q; Luo, X; Bang, S; Matsuoka, Y; McGinnis, A; Nackley, AG; Ji, R-R
Published in: Front Immunol
2021

The incidence of chronic pain is especially high in women, but the underlying mechanisms remain poorly understood. Interleukin-23 (IL-23) is a pro-inflammatory cytokine and contributes to inflammatory diseases (e.g., arthritis and psoriasis) through dendritic/T cell signaling. Here we examined the IL-23 involvement in sexual dimorphism of pain, using an optogenetic approach in transgenic mice expressing channelrhodopsin-2 (ChR2) in TRPV1-positive nociceptive neurons. In situ hybridization revealed that compared to males, females had a significantly larger portion of small-sized (100-200 μm2) Trpv1+ neurons in dorsal root ganglion (DRG). Blue light stimulation of a hindpaw of transgenic mice induced intensity-dependent spontaneous pain. At the highest intensity, females showed more intense spontaneous pain than males. Intraplantar injection of IL-23 (100 ng) induced mechanical allodynia in females only but had no effects on paw edema. Furthermore, intraplantar IL-23 only potentiated blue light-induced pain in females, and intrathecal injection of IL-23 also potentiated low-dose capsaicin (500 ng) induced spontaneous pain in females but not males. IL-23 expresses in DRG macrophages of both sexes. Intrathecal injection of IL-23 induced significantly greater p38 phosphorylation (p-p38), a marker of nociceptor activation, in DRGs of female mice than male mice. In THP-1 human macrophages estrogen and chemotherapy co-application increased IL-23 secretion, and furthermore, estrogen and IL-23 co-application, but not estrogen and IL-23 alone, significantly increased IL-17A release. These findings suggest a novel role of IL-23 in macrophage signaling and female-dominant pain, including C-fiber-mediated spontaneous pain. Our study has also provided new insight into cytokine-mediated macrophage-nociceptor interactions, in a sex-dependent manner.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

Front Immunol

DOI

EISSN

1664-3224

Publication Date

2021

Volume

12

Start / End Page

787565

Location

Switzerland

Related Subject Headings

  • p38 Mitogen-Activated Protein Kinases
  • TRPV Cation Channels
  • THP-1 Cells
  • Sex Characteristics
  • Pain Threshold
  • Pain
  • Optogenetics
  • Nociceptors
  • Nerve Fibers, Unmyelinated
  • Mice, Transgenic
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Ji, J., He, Q., Luo, X., Bang, S., Matsuoka, Y., McGinnis, A., … Ji, R.-R. (2021). IL-23 Enhances C-Fiber-Mediated and Blue Light-Induced Spontaneous Pain in Female Mice. Front Immunol, 12, 787565. https://doi.org/10.3389/fimmu.2021.787565
Ji, Jasmine, Qianru He, Xin Luo, Sangsu Bang, Yutaka Matsuoka, Aidan McGinnis, Andrea G. Nackley, and Ru-Rong Ji. “IL-23 Enhances C-Fiber-Mediated and Blue Light-Induced Spontaneous Pain in Female Mice.Front Immunol 12 (2021): 787565. https://doi.org/10.3389/fimmu.2021.787565.
Ji J, He Q, Luo X, Bang S, Matsuoka Y, McGinnis A, et al. IL-23 Enhances C-Fiber-Mediated and Blue Light-Induced Spontaneous Pain in Female Mice. Front Immunol. 2021;12:787565.
Ji, Jasmine, et al. “IL-23 Enhances C-Fiber-Mediated and Blue Light-Induced Spontaneous Pain in Female Mice.Front Immunol, vol. 12, 2021, p. 787565. Pubmed, doi:10.3389/fimmu.2021.787565.
Ji J, He Q, Luo X, Bang S, Matsuoka Y, McGinnis A, Nackley AG, Ji R-R. IL-23 Enhances C-Fiber-Mediated and Blue Light-Induced Spontaneous Pain in Female Mice. Front Immunol. 2021;12:787565.

Published In

Front Immunol

DOI

EISSN

1664-3224

Publication Date

2021

Volume

12

Start / End Page

787565

Location

Switzerland

Related Subject Headings

  • p38 Mitogen-Activated Protein Kinases
  • TRPV Cation Channels
  • THP-1 Cells
  • Sex Characteristics
  • Pain Threshold
  • Pain
  • Optogenetics
  • Nociceptors
  • Nerve Fibers, Unmyelinated
  • Mice, Transgenic