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

Front Immunol. 2021 Dec 7:12:787565. doi: 10.3389/fimmu.2021.787565. eCollection 2021.

Abstract

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.

Keywords: IL-23; dorsal root ganglion; macrophage; mechanical allodynia; nociceptor, optogenetics, spontaneous pain, sex dimorphism, TRPV1.

Publication types

  • Comparative Study
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Capsaicin
  • Channelrhodopsins / genetics
  • Channelrhodopsins / metabolism
  • Disease Models, Animal
  • Female
  • Ganglia, Spinal / drug effects*
  • Ganglia, Spinal / metabolism
  • Ganglia, Spinal / physiopathology
  • Humans
  • Interleukin-17 / metabolism
  • Interleukin-23 / toxicity*
  • Light
  • Macrophages / drug effects
  • Macrophages / metabolism
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Transgenic
  • Nerve Fibers, Unmyelinated / drug effects*
  • Nerve Fibers, Unmyelinated / metabolism
  • Nociceptors / drug effects*
  • Nociceptors / metabolism
  • Optogenetics
  • Pain / chemically induced*
  • Pain / genetics
  • Pain / metabolism
  • Pain / physiopathology
  • Pain Threshold / drug effects*
  • Sex Characteristics
  • THP-1 Cells
  • TRPV Cation Channels / genetics
  • TRPV Cation Channels / metabolism*
  • p38 Mitogen-Activated Protein Kinases / metabolism

Substances

  • Channelrhodopsins
  • IL17A protein, human
  • Interleukin-17
  • Interleukin-23
  • TRPV Cation Channels
  • TRPV1 protein, mouse
  • p38 Mitogen-Activated Protein Kinases
  • Capsaicin