Research report
Neuroblastoma Neuro2A cells stably expressing a cloned μ-opioid receptor: a specific cellular model to study acute and chronic effects of morphine

https://doi.org/10.1016/0169-328X(95)00014-JGet rights and content

Abstract

Several cellular systems display desensitization and downregulation of opioid receptors upon chronic treatment, suggesting that they could be used as a model system to understand opioid tolerance/ dependence. However, a model system containing a homogeneous population of μ-opioid receptors, the receptors at which morphine and related opioids act, has been lacking. To approach this problem, the μ-opioid receptor (MOR-1) was stably expressed in murine neuroblastoma Neuro2A cells after transfection. The expressed receptor was negatively coupled to adenylyl cyclase through Gi/Go proteins, displayed high affinity ligand binding, and was expressed in high number (2.06 pmol/mg of [3H]diprenorphine binding sites). In addition, loss of ability of μ-opioids to acutely inhibit forskolin-stimulated cAMP formation was observed after 4–24 h of chronic exposure to these agonists with concentrations as low as 300–500 nM. The effects of chronic morphine or [d-Ala2,N-MePhe4,Gly-ol]enkephalin (DAMGO) administration were found to be time- and concentration-dependent. Cross ‘tolerance’ was also observed. Thus the IC50 value of DAMGO to inhibit adenylyl cyclase was increased by 27-fold from 4.3 nM in control cells to 117 nM in cells pretreated with 300 nM morphine; there was no effect on the inhibition of adenylyl cyclase mediated by muscarinic receptors. Further, receptor downregulation accompanied the desensitization process. However, different time-dependence for these two processes suggests, in line with other studies, that these are entirely different cellular adaptation processes. In addition, the opioid antagonist naloxone induced an acute increase in intracellular cAMP level (2–3 times above the control level) following chronic agonist exposure. This process was also concentration-dependent. Overall, these results suggest that the cell line utilized in this study has a homogeneous population of μ-opioid receptors, providing an ideal cellular model to study the molecular mechanisms underlying chronic; morphine treatment.

References (35)

  • A. Tempel et al.

    Supersensitivity of brain opiate receptor subtypes after chronic naltrexone treatment

    Life Sci.

    (1982)
  • A.A. White et al.

    Improved two-step method for the assay of adenylate and guanylate cyclase

    Anal. Biochem.

    (1978)
  • E.L. Barker et al.

    5-Hydroxytryptamine, receptor density and mRNA levels in choroid plexus epithelial cells after treatment with mianserin and (−)-1-(4-bromo-2,5-di-methoxyphenyl)-2-aminopropane

    Mol. Pharmacol.

    (1993)
  • B.D. Carter et al.

    Receptor mechanisms of opioid tolerance in SHSY5Y human neural cells

    Mol. Pharmacol.

    (1993)
  • Chakrabarti, S., Prather, P.L., Yu, L., Law, P.Y. and Loh, H.H., Expression of the g-opioid receptor in CHO cells:...
  • K.J. Chang et al.

    Opioid peptides induce reduction of enkephalin receptors in cultured neuroblastoma cells

    Nature

    (1982)
  • C. Chen et al.

    Calcium phosphate-mediate gene transfer: a high efficient transfection system for stable transforming cells with plasmid DNA

    Mol. Cell. Biol.

    (1987)
  • Cited by (53)

    • Modulating μ-opioid receptor phosphorylation switches agonist-dependent signaling as reflected in PKCε activation and dendritic spine stability

      2011, Journal of Biological Chemistry
      Citation Excerpt :

      Morphine induces maximum ERK phosphorylation and adenylyl cyclase inhibition at 1 μm (20). Considering the relative affinity of the four agonists for OPRM1 and their abilities to induce maximum ERK phosphorylation and adenylyl cyclase inhibition (4, 22), DAMGO was used at 1 μm, whereas etorphine and fentanyl were used at 10 nm, to achieved equivalent concentrations. At these concentrations, the agonists induce ERK phosphorylation and adenylyl cyclase inhibition to similar levels.

    • Chronic morphine treatment up-regulates mu opioid receptor binding in cells lacking filamin A

      2007, Brain Research
      Citation Excerpt :

      Extensive studies in many different cell lines demonstrated MOP down-regulation by different agonists (Baumhaker et al., 1993; Chakrabarti et al., 1995; Kato et al., 1998; Yabaluri and Medzihradsky, 1997). Chronic morphine-induced MOP down-regulation has been well investigated and characterized in a various cell culture systems, such as SH-SY5Y (Zadina et al., 1993), HEK (Onoprishvili et al., 1999), CHO (Kato et al., 1998), C6 (Yabaluri and Medzihradsky, 1997), Neuro2a (Chakrabarti et al., 1995), 7315c (only 20%) (Puttfarcken and Cox, 1989) and SK-N-SH (Baumhaker et al., 1993). Opioid receptor up-regulation by antagonist treatment is a well-established phenomenon in animals and cell cultures.

    • Opioid abuse and brain gene expression

      2004, European Journal of Pharmacology
    • Turnover of μ-opioid receptors in neuroblastoma cells

      2002, Molecular Brain Research
      Citation Excerpt :

      N2A cells were cultured in DMEM supplemented with 10% fetal calf serum, 100 μg/ml streptomycin 100 IU/ml penicillin in a humidified atmosphere with 90% air and 10% CO2. N2A cells were transfected with constructs of MOR as previously described [5]. In order to facilitate the identification of the MOR with monoclonal antibody, a hemagglutinin (HA) epitope tag (YPYDVPDYA) recognized by the monoclonal antibody was spliced to the N-terminus immediately after the initial methionine codon as described earlier [2].

    View all citing articles on Scopus
    View full text