Elsevier

Neuroscience Letters

Volume 376, Issue 1, 7 March 2005, Pages 9-13
Neuroscience Letters

cAMP and protein kinase A contribute to the downregulation of spinal glutamate transporters after chronic morphine

https://doi.org/10.1016/j.neulet.2004.11.016Get rights and content

Abstract

Our previous study has shown that spinal glutamate transporters (GTs) are downregulated following chronic morphine administration; however, how spinal GTs are regulated in this process remains unclear. Here we show that the downregulation of spinal GTs (EAAC1 and GLT-1) induced by a 6-day intrathecal morphine (10 μg, twice daily) treatment regimen was prevented by co-administration of morphine with 2′,5′-dideoxyadenosine (ddA, 1 μg, a broad adenylyl cyclase inhibitor) or H89 (10 μg, a selective protein kinase A inhibitor). When co-administered with morphine, ddA or H89 also effectively attenuated the development of morphine tolerance in the same rats, while ddA or H89 alone did not affect the baseline nociceptive response. These results indicate that the downregulation of spinal GTs following chronic morphine is at least in part mediated through the intracellular cyclic AMP and protein kinase A pathway, suggesting that this cellular mechanism of GT regulation may be contributory to the development of morphine tolerance in rats.

Section snippets

Acknowledgements

This work was supported by PHS Grants RO1 DA08835, NS42661, and NS45681 to J.M.

References (35)

  • L.M. Bohn et al.

    Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence

    Nature

    (2000)
  • L.M. Bohn et al.

    Enhanced morphine analgesia in mice lacking beta-arrestin-2

    Science

    (1999)
  • F.E. D’Amour et al.

    A method for determining loss of pain sensation

    J. Pharmacol. Exp. Ther.

    (1941)
  • S.D. Ginsberg et al.

    Regional deafferentation downregulates subtypes of glutamate transporter protein

    J. Neurochem.

    (1995)
  • A.R. Gintzler et al.

    Opioid tolerance and the emergence of new opioid receptro-coupled signaling

    Mol. Neurobiol.

    (2001)
  • T. Jolas et al.

    Chronic morphine increases GABA tone on serotonergic neurons of the dorsal raphe nucleus: association with an up-regulation of the cyclic AMP pathway

    Neuroscience

    (2000)
  • J.C. Lievens et al.

    Characterization of striatal lesions produced by glutamate uptake alteration: cell death, reactive gliosis, and changes in GLT?1 and GADD45 mRNA expression

    Glia

    (2000)
  • Cited by (32)

    • The importance of the excitatory amino acid transporter 3 (EAAT3)

      2016, Neurochemistry International
      Citation Excerpt :

      Notably, chronic morphine treatment decreases EAAT3 expression through stimulation of opioid receptors in both rats, mice and cell lines (Mao et al., 2002). This was dependent on ubiquitination by NEDD4, an E3 ubiquitin ligase, which in turn was dependent on the phosphatase PTEN (Lim et al., 2005; Yang et al., 2008), though also EAAT3 mRNA decreases (Guo et al., 2015). The expression of EAAT3 was time-dependent, and after 12 h of withdrawal, the levels returned to baseline (Guo et al., 2015).

    • Effects of the altered activity of δ-opioid receptor on the expression of glutamate transporter type 3 induced by chronic exposure to morphine

      2013, Journal of the Neurological Sciences
      Citation Excerpt :

      The MOR agonist morphine was the focus of this study. Chronic morphine treatment has been shown to result in the down-regulation of Glu transporters (EAAT3 and EAAT2) in the spinal cord in rats, which is mediated (at least in part) by adenylate cyclase and PKA [11]. Yang et al. found that down-regulation of EAAT3 expression and decreased function caused by chronic morphine exposure could be blocked in C6 cells by the protease inhibitor MG-132, suggesting that the ubiquitin–proteasome system (UPS) is involved in this process.

    • Role of neuroinflammation in morphine tolerance: Effect of tumor necrosis factor-α

      2012, Acta Anaesthesiologica Taiwanica
      Citation Excerpt :

      Glutamatergic receptors, in particular NMDA receptors, are critically involved in chronic opioid-induced neuronal adaptations, such as opioid tolerance, dependence, and withdrawal,10,48,49 and in chronic pain-associated hyperalgesia.50–52 Emerging evidence suggests that opioid tolerance and abnormal pain sensitivity, the opioid-induced hyperalgesia, may share common cellular mechanisms and mediate, at least in part, through NMDA receptors12,53 and GT system.54 Our previous studies have demonstrated that coinfusion of the NMDA antagonist MK-801 with morphine shifted the AD50 (analgesic dose) of morphine to 11.2 μg compared to 83.8 μg in morphine tolerant-rats.55

    View all citing articles on Scopus
    View full text