Intrathecal delivery of a mutant micro-opioid receptor activated by naloxone as a possible antinociceptive paradigm

J Pharmacol Exp Ther. 2010 Sep 1;334(3):739-45. doi: 10.1124/jpet.109.165399. Epub 2010 Jun 16.

Abstract

Direct injection of double-stranded adeno-associated virus type 2 (dsAAV2) with a mu-opioid receptor (MOR) mutant [S4.45(196)A], and a reporter protein (enhanced green fluorescent protein) into the spinal cord (S2/S3) dorsal horn region of ICR mice resulted in antinociceptive responses to systemic injection of opioid antagonist naloxone without altering the acute agonist morphine responses and no measurable tolerance or dependence development during subchronic naloxone treatment. To develop further such mutant MORs into a therapeutic agent in pain management, a less invasive method for virus delivery is needed. Thus, in current studies, the dsAAV2 was locally injected into the subarachnoid space of the spinal cord by intrathecal administration. Instead of using the MORS196A mutant, we constructed the dsAAV2 vector with the MORS196ACSTA mutant, a receptor mutant in which naloxone has been shown to exhibit full agonistic properties in vitro. After 2 weeks of virus injection, naloxone (10 mg/kg s.c.) elicited antinociceptive effect (determined by tail-flick test) without tolerance (10 mg/kg s.c., b.i.d. for 6 days) and significant withdrawal symptoms. On the other hand, subchronic treatment with morphine (10 mg/kg s.c., b.i.d.) for 6 days induced significant tolerance (4.8-fold) and withdrawal symptoms. Furthermore, we found that morphine, but not naloxone, induced the rewarding effects (determined by conditioned place preference test). These data suggest that local expression of MORS196ACSTA in spinal cord and systemic administration of naloxone has the potential to be developed into a new strategy in the management of pain without addiction liability.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Conditioning, Operant / drug effects
  • Drug Tolerance
  • Gene Transfer Techniques
  • Genetic Therapy
  • Injections, Spinal
  • Male
  • Mice
  • Mice, Inbred ICR
  • Mutation
  • Naloxone / pharmacology*
  • Narcotic Antagonists / pharmacology*
  • Opioid-Related Disorders / psychology
  • Pain Management*
  • Pain Measurement / drug effects*
  • Receptors, Opioid, mu / drug effects*
  • Receptors, Opioid, mu / genetics*
  • Reward
  • Subarachnoid Space

Substances

  • Narcotic Antagonists
  • Receptors, Opioid, mu
  • Naloxone