Inhibition of microglial P2X4 receptors attenuates morphine tolerance, Iba1, GFAP and mu opioid receptor protein expression while enhancing perivascular microglial ED2

Pain. 2010 Sep;150(3):401-413. doi: 10.1016/j.pain.2010.02.042. Epub 2010 Jun 22.

Abstract

Anti-nociceptive tolerance to opioids is a well-described phenomenon, which severely limits the clinical efficacy of opioids for the treatment of chronic pain syndromes. The mechanisms that drive anti-nociceptive tolerance, however, are less well understood. We have previously shown that glia have a central role in the development of morphine tolerance and that administration of a glial modulating agent attenuated tolerance formation. Recently, we have demonstrated that morphine enhances microglial Iba1 expression and P2X4 receptor-mediated microglial migration via direct mu opioid receptor signaling in in vitro microglial cultures. We hypothesize that P2X4 receptors drive morphine tolerance and modulate morphine-induced spinal glial reactivity. Additionally, we hypothesize that perivascular microglia play a role in morphine tolerance and that P2X4 receptor expression regulates perivascular microglia ED2 expression. To test these hypotheses, rats were implanted with osmotic minipumps releasing morphine or saline subcutaneously for seven days. Beginning three days prior to morphine treatment, P2X4 receptor antisense oligonucleotide (asODN) was injected intrathecally daily, to selectively inhibit P2X4 receptor expression. P2X4 receptor asODN treatment inhibited morphine-induced P2X4 receptor expression and blocked anti-nociceptive tolerance to systemically administered morphine. P2X4 receptor asODN treatment also attenuated the morphine-dependent increase of spinal ionized calcium binding protein (Iba1), glial fibrillary acidic protein (GFAP) and mu opioid receptor protein expression. Chronic morphine also decreased perivascular microglial ED2 expression, which was reversed by P2X4 receptor asODN. Together, these data suggest that the modulation of P2X4 receptor expression on microglia and perivascular microglia may prove an attractive target for adjuvant therapy to attenuate opioid-induced anti-nociceptive tolerance.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Analysis of Variance
  • Animals
  • Antigens, CD / metabolism
  • Antigens, Differentiation, Myelomonocytic / metabolism
  • Behavior, Animal / drug effects
  • CD11b Antigen / metabolism
  • Calcium-Binding Proteins / metabolism*
  • Disease Models, Animal
  • Drug Interactions
  • Drug Tolerance / physiology*
  • Gene Expression Regulation / drug effects
  • Gene Expression Regulation / physiology*
  • Glial Fibrillary Acidic Protein / metabolism
  • Hyperalgesia / drug therapy
  • Male
  • Microfilament Proteins
  • Microglia / drug effects
  • Microglia / metabolism*
  • Morphine / administration & dosage*
  • Naloxone / pharmacology
  • Narcotic Antagonists / pharmacology
  • Nerve Tissue Proteins / metabolism
  • Oligodeoxyribonucleotides, Antisense / pharmacology
  • Pain Measurement
  • Random Allocation
  • Rats
  • Rats, Sprague-Dawley
  • Receptors, Cell Surface / metabolism
  • Receptors, Opioid / metabolism*
  • Receptors, Purinergic P2X4 / genetics
  • Receptors, Purinergic P2X4 / metabolism*
  • Signal Transduction
  • Spinal Cord / pathology
  • Time Factors

Substances

  • Aif1 protein, rat
  • Antigens, CD
  • Antigens, Differentiation, Myelomonocytic
  • CD11b Antigen
  • CD163 antigen
  • Calcium-Binding Proteins
  • Glial Fibrillary Acidic Protein
  • Microfilament Proteins
  • Narcotic Antagonists
  • Nerve Tissue Proteins
  • Oligodeoxyribonucleotides, Antisense
  • Receptors, Cell Surface
  • Receptors, Opioid
  • Receptors, Purinergic P2X4
  • Naloxone
  • Morphine