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

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. doi: 10.1016/j.neuron.2021.06.015.
Xin Luo 1 Ouyang Chen 2 Zilong Wang 3 Sangsu Bang 3 Jasmine Ji 3 Sang Hoon Lee 4 Yul Huh 2 Kenta Furutani 3 Qianru He 3 Xueshu Tao 3 Mei-Chuan Ko 5 Andrey Bortsov 3 Christopher R Donnelly 3 Yong Chen 6 Andrea Nackley 7 Temugin Berta 4 Ru-Rong Ji 8
Affiliations

Affiliations

  • 1 Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA. Electronic address: [email protected].
  • 2 Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
  • 3 Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
  • 4 Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
  • 5 Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
  • 6 Department of Neurology, Duke University Medical Center, Durham, NC, USA.
  • 7 Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
  • 8 Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC, USA. Electronic address: [email protected].
Abstract

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.

Keywords

IL-17; IL-23; dorsal root ganglion; estrogen receptor α; human; macrophage; mechanical allodynia; nociceptor; nonhuman primate; sex dimorphism.

Figures
Products