Elsevier

Neuropharmacology

Volume 60, Issue 1, January 2011, Pages 58-65
Neuropharmacology

The role of beta-arrestin2 in the severity of antinociceptive tolerance and physical dependence induced by different opioid pain therapeutics

https://doi.org/10.1016/j.neuropharm.2010.08.003Get rights and content

Abstract

Ligands acting at the same receptor can differentially activate distinct signal transduction pathways, which in turn, can have diverse functional consequences. Further, receptors expressed in different tissues may utilize intracellular signaling proteins in response to a ligand differently as well. The mu opioid receptor (MOR), which mediates many of the pharmacological actions of opiate therapeutics, is also subject to differential signaling in response to diverse agonists. To study the effect of diverse agonists on MOR signaling, we examined the effects of chronic opiate treatment on two distinct physiological endpoints, antinociceptive tolerance and physical dependence, in mice lacking the intracellular regulatory molecule, βarrestin2. While βarrestin2 knockout (βarr2-KO) mice do not become tolerant to the antinociceptive effects of chronic morphine in a hot plate test, tolerance develops to the same degree in both wild type and βarr2-KO mice following chronic infusion with methadone, fentanyl, and oxycodone. Studies here also assess the severity of withdrawal signs precipitated by naloxone following chronic infusions at three different doses of each opiate agonist. While there are no differences in withdrawal responses between genotypes at the highest dose of morphine tested (48 mg/kg/day), the βarr2-KO mice display several less severe withdrawal responses when the infusion dose is lowered (12 or 24 mg/kg/day). Chronic infusion of methadone, fentanyl, and oxycodone all lead to equivalent naloxone-precipitated withdrawal responses in both genotypes at all doses tested. These results lend further evidence that distinct agonists can differentially impact on opioid-mediated responses in vivo in a βarrestin2-dependent manner.

Introduction

Opioid analgesics are commonly used to treat moderate to severe pain. Long-term administration, however, is associated with the development of undesirable side effects including analgesic tolerance and physical dependence. A number of studies have demonstrated that the physiological actions of morphine and other clinically used opiates are mediated primarily through activation of the mu opioid receptor (MOR), a G protein-coupled receptor (Matthes et al., 1996, Sora et al., 1997, Roy et al., 1998, Kieffer, 1999). While diverse signaling components and complex neuronal adaptations contribute to the development of analgesic tolerance and physical dependence, considerable evidence suggests that regulation of MOR signaling can affect these adaptive responses.

Previously, we have shown that βarrestin2, an important G protein-coupled receptor regulatory protein, differentially regulates opiate effects in a manner that is determined by the agonist. In βarrestin2 knockout (βarr2-KO) mice, acute morphine and heroin-induced antinociception is enhanced and prolonged, while acute antinociceptive responses to etorphine, methadone, and fentanyl are similar to those observed in their wild type (WT) counterparts (Bohn et al., 1999, Bohn et al., 2004). Further, βarr2-KO mice also do not develop antinociceptive tolerance in a hot plate test following treatment with a single high dose of morphine or after chronic treatment using either repeated injections of morphine for 9 days or implantation with a 75 mg morphine pellet for 3 days (Bohn et al., 2000, Bohn et al., 2002). However, the extent of physical dependence produced following a 72 h treatment with the 75 mg morphine pellet is equivalent between both WT and βarr2-KO mice (Bohn et al., 2000). These earlier studies demonstrate a complex role for βarrestin2 in the regulation of morphine-induced antinociceptive tolerance and physical dependence; however, its role in MOR regulation with other agonists following chronic administration has not been previously investigated.

In this study, we have evaluated the development of tolerance and dependence in WT and βarr2-KO mice following chronic opiate infusion using implanted osmotic pumps. Since patients suffering from moderate to severe pain are commonly treated with sustained released formulations of morphine (MS Contin®), fentanyl (Duragesic®), and oxycodone (Oxycontin®), a continuous drug infusion paradigm used in these studies may closely mimic the exposure to opiate drugs in a clinical setting. Furthermore, while prior studies suggest that βarrestin2 does not play a significant role in morphine-induced physical dependence or in the display of somatic withdrawal signs, morphine was administered using a 75 mg pellet implantation in those studies (Bohn et al., 2000). Since this high dosing regimen may have occluded βarrestin2 contributions to this adaptive response, the effects of lower doses of morphine, as well as several doses of methadone, fentanyl and oxycodone, are assessed in this current study.

Section snippets

Animals

Male WT and βarr2-KO mice were generated from heterozygous breeding as previously described (Bohn et al., 1999). Mice were age matched (3–6 months old), weighed between 20 and 35 g, were group housed in a temperature-controlled room, and were maintained on a 12 h reversed light/dark cycle. All behavioral studies were conducted during the light phase of the animal’s circadian cycle. Mice had free access to food and water prior to experiments. Both genotypes were tested in parallel and each mouse

Results

Antinociceptive tolerance in WT and βarr2-KO mice was evaluated using two different treatment paradigms. In the first paradigm, mice were assessed for their response latencies to a thermal hot plate stimulus every other day during 5 days of chronic agonist treatment. While the half-life of morphine is approximately 2 h in mice (Aceto et al., 1997), the half-lives of methadone, fentanyl, and oxycodone are much shorter at 60, 45 and 30 min respectively (Duttaroy and Yoburn, 1995, Kalvass et al.,

Discussion

In this study several different opioid analgesics were compared for their ability to induce antinociceptive tolerance and physical dependence in WT and βarr2-KO mice following chronic infusion via osmotic pumps. Under these dosing conditions, morphine produces robust tolerance in WT mice, while this effect is greatly attenuated in the βarr2-KO mice. At high doses of morphine both WT and βarr2-KO mice display equivalent somatic signs of withdrawal, while at lower doses, the βarr2-KO mice display

Acknowledgements

This work was supported by the National Institute on Drug Abuse grants DA021952 (K.M.R), DA14600 and DA18860 (L.M.B.)

References (44)

  • K.M. Raehal et al.

    Morphine-induced physiological and behavioral responses in mice lacking G protein-coupled receptor kinase 6

    Drug Alcohol Depend.

    (2009)
  • S. Roy et al.

    mu-Opioid receptor-knockout mice: the role of mu-opioid receptor in gastrointestinal transit

    Brain Res. Mol. Brain Res.

    (1998)
  • C.L. Schmid et al.

    Physiological and pharmacological implications of beta-arrestin regulation

    Pharmacol. Ther.

    (2009)
  • F.L. Smith et al.

    PKC and PKA inhibitors reinstate morphine-induced behaviors in morphine tolerant mice

    Pharmacol. Res.

    (2006)
  • A. Tjolsen et al.

    An improved method for tail-flick testing with adjustment for tail-skin temperature

    J. Neurosci. Methods

    (1989)
  • J.L. Whistler et al.

    Functional dissociation of mu opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and addiction

    Neuron

    (1999)
  • B.C. Yoburn et al.

    Supersensitivity to opioid analgesics following chronic opioid antagonist treatment: relationship to receptor selectivity

    Pharmacol. Biochem. Behav.

    (1995)
  • M.D. Aceto et al.

    Dependence studies of new compounds in the rhesus monkey, rat and mouse

  • J.R. Arden et al.

    Phosphorylation and agonist-specific intracellular trafficking of an epitope-tagged mu-opioid receptor expressed in HEK 293 cells

    J. Neurochem.

    (1995)
  • S. Arttamangkul et al.

    Differential activation and trafficking of micro-opioid receptors in brain slices

    Mol. Pharmacol.

    (2008)
  • F. Berrendero et al.

    Increase of morphine withdrawal in mice lacking A2a receptors and no changes in CB1/A2a double knockout mice

    Eur. J. Neurosci.

    (2003)
  • L.M. Bohn et al.

    Relative opioid efficacy is determined by the complements of the G protein-coupled receptor desensitization machinery

    Mol. Pharmacol.

    (2004)
  • Cited by (156)

    View all citing articles on Scopus
    View full text