1. Academic Validation
  2. TRPV1 promotes opioid analgesia during inflammation

TRPV1 promotes opioid analgesia during inflammation

  • Sci Signal. 2019 Apr 2;12(575):eaav0711. doi: 10.1126/scisignal.aav0711.
Lilian Basso 1 Reem Aboushousha 1 Churmy Yong Fan 2 Mircea Iftinca 1 Helvira Melo 1 Robyn Flynn 1 Francina Agosti 3 Morley D Hollenberg 1 Roger Thompson 2 Emmanuel Bourinet 3 Tuan Trang 2 Christophe Altier 4
Affiliations

Affiliations

  • 1 Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada.
  • 2 Hotchkiss Brain Institute, Cumming School of Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada.
  • 3 Institute for Functional Genomics, CNRS UMR5203, INSERM U1191, University of Montpellier, LABEX ICST, Montpellier, France.
  • 4 Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada. altier@ucalgary.ca.
Abstract

Pain and inflammation are inherently linked responses to injury, Infection, or chronic diseases. Given that acute inflammation in humans or mice enhances the analgesic properties of opioids, there is much interest in determining the inflammatory transducers that prime Opioid Receptor signaling in primary afferent nociceptors. Here, we found that activation of the transient receptor potential vanilloid type 1 (TRPV1) channel stimulated a mitogen-activated protein kinase (MAPK) signaling pathway that was accompanied by the shuttling of the scaffold protein β-arrestin2 to the nucleus. The nuclear translocation of β-arrestin2 in turn prevented its recruitment to the μ-opioid receptor (MOR), the subsequent internalization of agonist-bound MOR, and the suppression of MOR activity that occurs upon receptor desensitization. Using the complete Freund's adjuvant (CFA) inflammatory pain model to examine the role of TRPV1 in regulating endogenous opioid analgesia in mice, we found that naloxone methiodide (Nal-M), a peripherally restricted, nonselective, and competitive Opioid Receptor antagonist, slowed the recovery from CFA-induced hypersensitivity in wild-type, but not TRPV1-deficient, mice. Furthermore, we showed that inflammation prolonged morphine-induced antinociception in a mouse model of Opioid Receptor desensitization, a process that depended on TRPV1. Together, our data reveal a TRPV1-mediated signaling pathway that serves as an endogenous pain-resolution mechanism by promoting the nuclear translocation of β-arrestin2 to minimize MOR desensitization. This previously uncharacterized mechanism may underlie the peripheral opioid control of inflammatory pain. Dysregulation of the TRPV1-β-arrestin2 axis may thus contribute to the transition from acute to chronic pain.

Figures
Products