Your browser doesn't support javascript.
loading
Site-Specific Regulation of P2X7 Receptor Function in Microglia Gates Morphine Analgesic Tolerance.
Leduc-Pessah, Heather; Weilinger, Nicholas L; Fan, Churmy Y; Burma, Nicole E; Thompson, Roger J; Trang, Tuan.
Affiliation
  • Leduc-Pessah H; Departments of Comparative Biology & Experimental Medicine, and Physiology & Pharmacology.
  • Weilinger NL; Hotchkiss Brain Institute, and.
  • Fan CY; Hotchkiss Brain Institute, and.
  • Burma NE; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
  • Thompson RJ; Departments of Comparative Biology & Experimental Medicine, and Physiology & Pharmacology.
  • Trang T; Hotchkiss Brain Institute, and.
J Neurosci ; 37(42): 10154-10172, 2017 10 18.
Article in En | MEDLINE | ID: mdl-28924009
ABSTRACT
Tolerance to the analgesic effects of opioids is a major problem in chronic pain management. Microglia are implicated in opioid tolerance, but the core mechanisms regulating their response to opioids remain obscure. By selectively ablating microglia in the spinal cord using a saporin-conjugated antibody to Mac1, we demonstrate a causal role for microglia in the development, but not maintenance, of morphine tolerance in male rats. Increased P2X7 receptor (P2X7R) activity is a cardinal feature of microglial activation, and in this study we found that morphine potentiates P2X7R-mediated Ca2+ responses in resident spinal microglia acutely isolated from morphine tolerant rats. The increased P2X7R function was blocked in cultured microglia by PP2, a Src family protein tyrosine kinase inhibitor. We identified Src family kinase activation mediated by µ-receptors as a key mechanistic step required for morphine potentiation of P2X7R function. Furthermore, we show by site-directed mutagenesis that tyrosine (Y382-384) within the P2X7R C-terminus is differentially modulated by repeated morphine treatment and has no bearing on normal P2X7R function. Intrathecal administration of a palmitoylated peptide corresponding to the Y382-384 site suppressed morphine-induced microglial reactivity and preserved the antinociceptive effects of morphine in male rats. Thus, site-specific regulation of P2X7R function mediated by Y382-384 is a novel cellular determinant of the microglial response to morphine that critically underlies the development of morphine analgesic tolerance.SIGNIFICANCE STATEMENT Controlling pain is one of the most difficult challenges in medicine and its management is a requirement of a large diversity of illnesses. Although morphine and other opioids offer dramatic and impressive relief of pain, their impact is truncated by loss of efficacy (analgesic tolerance). Understanding why this occurs and how to prevent it are of critical importance in improving pain therapies. We uncovered a novel site (Y382-384) within the P2X7 receptor that can be targeted to blunt the development of morphine analgesic tolerance, without affecting normal P2X7 receptor function. Our findings provide a critical missing mechanistic piece, site-specific modulation by Y382-384, that unifies P2X7R function to the activation of spinal microglia and the development of morphine tolerance.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pain Measurement / Microglia / Receptors, Purinergic P2X7 / Analgesics, Opioid / Morphine Type of study: Prognostic_studies Limits: Animals Language: En Journal: J Neurosci Year: 2017 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pain Measurement / Microglia / Receptors, Purinergic P2X7 / Analgesics, Opioid / Morphine Type of study: Prognostic_studies Limits: Animals Language: En Journal: J Neurosci Year: 2017 Document type: Article