Review
Allostatic Mechanisms of Opioid Tolerance Beyond Desensitization and Downregulation

https://doi.org/10.1016/j.tips.2016.08.002Get rights and content

Trends

Behavioral tolerance to opioids results from many interacting processes including desensitization, resensitization, and cellular and pass-forward allostasis.

Opioid-biased agonism presents new pharmacological approaches to avoid detrimental side effects and negate aspects of opioid tolerance and dependence.

Negative affect is an opponent process that creates behavioral tolerance to the rewarding effects of opioid drugs, and constitutes a likely driver of relapse following drug cessation.

Microglia activation following chronic opioids is a potential therapeutic target contributing to behavioral tolerance and dependence mechanisms.

Mechanisms of opioid tolerance have focused on adaptive modifications within cells containing opioid receptors, defined here as cellular allostasis, emphasizing regulation of the opioid receptor signalosome. We review additional regulatory and opponent processes involved in behavioral tolerance, and include mechanistic differences both between agonists (agonist bias), and between μ- and δ-opioid receptors. In a process we will refer to as pass-forward allostasis, cells modified directly by opioid drugs impute allostatic changes to downstream circuitry. Because of the broad distribution of opioid systems, every brain cell may be touched by pass-forward allostasis in the opioid-dependent/tolerant state. We will implicate neurons and microglia as interactive contributors to the cumulative allostatic processes creating analgesic and hedonic tolerance to opioid drugs.

Section snippets

What is Tolerance?

Tolerance (see Glossary) is defined as a reduction in effect following prolonged drug administration that results in a loss of drug potency indicated by a pharmacological shift to the right in the dose–response curve. The development and extent of tolerance are dependent on the drug interactions with the opioid receptor(s), dose, and frequency of administration. There are many mechanisms that can contribute to opioid tolerance at a behavioral level, including upregulation of drug metabolism

Cellular Allostasis and the Opioid Receptor Signalosome (MOP and DOP)

Mechanisms of opioid receptor desensitization and resensitization have been thoroughly reviewed for different agonists at the MOP [13]. The ability of different agonists to induce receptor internalization has been shown to affect divergent downstream regulatory processes contributing to the activity of the opioid receptor signalosome and cellular tolerance (Figure 1). Many other processes contribute to cellular allostasis. MOP signaling initiates a cascade of cellular adaptations involving

The Complex Role of Arrestins in Behavioral Tolerance to Opioids

Following the initial report that mutant mice lacking arrestin 3 (β-arrestin 2) exhibit attenuated tolerance to morphine [18], there is now a plethora of studies demonstrating the involvement of arrestins in opioid receptor desensitization and tolerance. Both high- and low-internalizing MOP and DOP agonists induce analgesic tolerance 19, 20, 21, 22, albeit via different mechanisms. Moreover, MOP receptor agonists, independently of their internalization efficacy, exhibit comparable symptoms of

Opponent Processes Contribute to a Tolerant State

Behavioral opioid tolerance is most often studied in the context of analgesia and cellular signaling mechanisms, as reviewed above. However, opponent processes are also evident at a circuit-level in the regulation of mood and affect. In this section we will focus on how chronic opioid exposure causes adaptations of the mesocorticolimbic circuitry as a result of the development of opponent processes in the striatum and ventral tegmental area (VTA). The striatum is an important hub of the opioid

Allostatic Processes in the VTA

Dopamine release from VTA dopaminergic neurons reinforces natural rewarding behavior and attributes motivational salience to otherwise neutral environmental stimuli or unanticipated salient stimuli 34, 37, 38. Dopaminergic neurons from the VTA exhibit a rich and complex organization, with widespread projections to forebrain targets 39, 40, 41, 42, 43. The rewarding effects of morphine are mediated via activation of MOP because mice lacking this receptor do not express conditioned place

Allostatic Processes in Striatal Cells, Signaling, and Circuits

Similarly to other drugs of abuse, chronic morphine induces extensive and region-specific synaptic plasticity within the striatum. Glutamate homeostasis is affected by changes in glutamate release and reuptake, and in the composition, number, and role of ionotropic and metabotropic receptors ([72] for review). At the level of the synapse, a 10 day period of withdrawal from chronic non-contingent (i.e., experimenter-delivered) morphine increased synaptic strength, as assessed by the ratio of

Opioid-Mediated Aversion Circuitry

The negative affect and ‘anti-reward’ that is created by an opioid-dependent state is now recognized as a driving contributor to drug-seeking behaviors 85, 86, 87, and the neurocircuitry of the learned association between drug-induced relief of these aversive states may be particularly important for opioid substance-abuse disorder. Repeated or chronic use of opioids induces adaptive or allostatic changes that modify neuronal circuitry and create an altered normality – the ‘drug-dependent’ state

Concluding Remarks and Future Directions

Tolerance to opioids per se is not a reliable predictor of abuse liability but presents a challenge for treating both acute and chronic pain, and becomes a liability for addicts that relapse. Opioid tolerance is also a predictor of significantly longer length of hospital stay and readmission rates. Tolerance often masks opioid-induced hyperalgesia and contributes to the development of affective disorders such as anxiety and depression, where the emergence of anxiety during withdrawal can be

Acknowledgments

Financial support was provided by the Shirley and Stefan Hatos Foundation (C.J.E., A.M.W.T., W.W)., Shirley Hatos (C.M.C.), Cousins Center for Psychoneuroimmunology (AMWT), National Institutes of Health (NIH) K99DA040016 (A.M.W.T.), NIH DA005010 (C.J.E., A.M.W.T., W.W.), NIH DA031243 (A.A.A.P.), Department of Defense MR141282 (C.M.C.), and the University of Illinois at Chicago Department of Psychiatry (A.A.A.P.).

Glossary

Allostasis
adaptive modifications of the nervous system following chronic opioid use that create a new stable state dependent on presence of the drug.
Brain-derived neurotrophic factor (BDNF)
factor released from neurons and/or microglia that has been shown to contribute to neuronal plasticity during development and pathology, including chronic opioid exposure.
Long-term depression (LTD)
an activity-dependent decrease in the strength (or efficacy) of a neuronal synapse that can last for hours or

References (128)

  • J.P. Britt

    Synaptic and behavioral profile of multiple glutamatergic inputs to the nucleus accumbens

    Neuron

    (2012)
  • K.C. Berridge et al.

    What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience?

    Brain Res. Brain Res. Rev.

    (1998)
  • M. Pignatelli et al.

    Role of dopamine neurons in reward and aversion: a synaptic plasticity perspective

    Neuron

    (2015)
  • H.L. Fields et al.

    Understanding opioid reward

    Trends Neurosci.

    (2015)
  • A. Matsui

    Separate GABA afferents to dopamine neurons mediate acute action of opioids, development of tolerance, and expression of withdrawal

    Neuron

    (2014)
  • A. Bechara

    A two-separate-motivational-systems hypothesis of opioid addiction

    Pharmacol. Biochem. Behav.

    (1998)
  • S.R. Laviolette

    Motivational state determines the functional role of the mesolimbic dopamine system in the mediation of opiate reward processes

    Behav. Brain Res.

    (2002)
  • N.A. Crowley

    Dynorphin controls the gain of an amygdalar anxiety circuit

    Cell Rep.

    (2016)
  • N. Doyon

    Chloride regulation: a dynamic equilibrium crucial for synaptic inhibition

    Neuron

    (2016)
  • X. Wu

    Effects of morphine withdrawal on the membrane properties of medium spiny neurons in the nucleus accumbens shell

    Brain Res. Bull.

    (2013)
  • B. Ziolkowska

    Temporal and anatomic patterns of immediate-early gene expression in the forebrain of C57BL/6 and DBA/2 mice after morphine administration

    Neuroscience

    (2015)
  • T. Enoksson

    Nucleus accumbens D2- and D1-receptor expressing medium spiny neurons are selectively activated by morphine withdrawal and acute morphine, respectively

    Neuropharmacology

    (2012)
  • G.F. Koob

    Addiction as a stress surfeit disorder

    Neuropharmacology

    (2014)
  • G.F. Koob

    Negative reinforcement in drug addiction: the darkness within

    Curr. Opin. Neurobiol.

    (2013)
  • F. Papaleo

    Disruption of the CRF/CRF1 receptor stress system exacerbates the somatic signs of opiate withdrawal

    Neuron

    (2007)
  • N. Baharlouei

    Blockage of acquisition and expression of morphine-induced conditioned place preference in rats due to activation of glutamate receptors type II/III in nucleus accumbens

    Pharmacol. Biochem. Behav.

    (2015)
  • A. Haghparast

    Cannabinoid receptors in the basolateral amygdala are involved in the potentiation of morphine rewarding properties in the acquisition, but not expression of conditioned place preference in rats

    Brain Res.

    (2014)
  • P. Gulur

    Opioid tolerance – a predictor of increased length of stay and higher readmission rates

    Pain Physician

    (2014)
  • N.D. Volkow et al.

    Opioid abuse in chronic pain – misconceptions and mitigation strategies

    N. Engl. J. Med.

    (2016)
  • S. Siegel

    Heroin ‘overdose’ death: contribution of drug-associated environmental cues

    Science

    (1982)
  • T.W. Vanderah

    Delta and kappa opioid receptors as suitable drug targets for pain

    Clin. J. Pain

    (2010)
  • L. Gendron

    Molecular pharmacology of delta-opioid receptors

    Pharmacol. Rev.

    (2016)
  • A. Charles et al.

    Delta-opioid receptors as targets for migraine therapy

    Curr. Opin. Neurol.

    (2016)
  • C. Chavkin et al.

    Kappa antagonist JDTic in Phase 1 clinical trial

    Neuropsychopharmacology

    (2015)
  • L. Toll

    Nociceptin/orphanin FQ receptor structure, signaling, ligands, functions, and interactions with opioid systems

    Pharmacol. Rev.

    (2016)
  • A.A. Pradhan

    Ligand-directed signalling within the opioid receptor family

    Br. J. Pharmacol.

    (2012)
  • J.T. Williams

    Regulation of mu-opioid receptors: desensitization, phosphorylation, internalization, and tolerance

    Pharmacol. Rev.

    (2013)
  • R. Al-Hasani et al.

    Molecular mechanisms of opioid receptor-dependent signaling and behavior

    Anesthesiology

    (2011)
  • H. Sugiura

    Transducing neuronal activity into dendritic spine morphology: new roles for p38 MAP kinase and N-cadherin

    Neuroscientist

    (2009)
  • M.S. Mazei-Robison et al.

    Opiate-induced molecular and cellular plasticity of ventral tegmental area and locus coeruleus catecholamine neurons

    Cold Spring Harb. Perspect. Med.

    (2012)
  • L. Zhang

    A novel noncanonical signaling pathway for the mu-opioid receptor

    Mol. Pharmacol.

    (2013)
  • L.M. Bohn

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

    Nature

    (2000)
  • A.A. Pradhan

    Agonist-specific recruitment of arrestin isoforms differentially modify delta opioid receptor function

    J. Neurosci.

    (2016)
  • E.A. Walker et al.

    Differential tolerance to antinociceptive effects of mu opioids during repeated treatment with etonitazene, morphine, or buprenorphine in rats

    Psychopharmacology

    (2001)
  • A.A. Pradhan

    Ligand-directed trafficking of the delta-opioid receptor in vivo: two paths toward analgesic tolerance

    J. Neurosci.

    (2010)
  • C. Xu

    Serine 363 of the δ-opioid receptor is crucial for adopting distinct pathways to activate ERK1/2 in response to stimulation with different ligands

    J. Cell Sci.

    (2010)
  • N. Mittal

    Evidence that behavioral phenotypes of morphine in beta-arr2-/- mice are due to the unmasking of JNK signaling

    Neuropsychopharmacology

    (2012)
  • C.G. Pontrello

    Cofilin under control of beta-arrestin-2 in NMDA-dependent dendritic spine plasticity, long-term depression (LTD), and learning

    Proc. Natl. Acad. Sci. U.S.A.

    (2012)
  • W. Walwyn

    Beta-arrestin2 and c-Src regulate the constitutive activity and recycling of mu opioid receptors in dorsal root ganglion neurons

    J. Neurosci.

    (2007)
  • K.M. Raehal

    Morphine side effects in beta-arrestin 2 knockout mice

    J. Pharmacol. Exp. Ther.

    (2005)
  • Cited by (85)

    • Opioid-induced microglia reactivity modulates opioid reward, analgesia, and behavior

      2022, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      As with TNFα and IL-1β, evidence also suggests that BDNF inhibits the rewarding effects of opioids. As mentioned earlier, elevated levels of BDNF disrupts chloride ion homeostasis in such a way that GABAergic activity is increased (Cahill et al., 2016; Ferrini et al., 2013; Koo et al., 2012; Taylor et al., 2016; Vargas-Perez et al., 2009, 2014). Importantly, it has been shown that opioid-dependent rats have increased levels of BDNF in the VTA (150 % greater than in controls) (Vargas-Perez et al., 2009).

    View all citing articles on Scopus
    View full text