Review
Opioid receptors: distinct roles in mood disorders

https://doi.org/10.1016/j.tins.2012.11.002Get rights and content

The roles of opioid receptors in pain and addiction have been extensively studied, but their function in mood disorders has received less attention. Accumulating evidence from animal research reveals that mu, delta and kappa opioid receptors (MORs, DORs and KORs, respectively) exert highly distinct controls over mood-related processes. DOR agonists and KOR antagonists have promising antidepressant potential, whereas the risk–benefit ratio of currently available MOR agonists as antidepressants remains difficult to evaluate, in addition to their inherent abuse liability. To date, both human and animal studies have mainly examined MORs in the etiology of depressive disorders, and future studies will address DOR and KOR function in established and emerging neurobiological aspects of depression, including neurogenesis, neurodevelopment, and social behaviors.

Introduction

Endogenous opioids and their receptors were discovered from the early use of opium in both medicinal and recreational practices. Morphine, the most active ingredient of opium, has highly potent pain-relieving properties and remains the most widely used painkiller in modern medicine, despite a plethora of undesirable effects. Among these, abuse liability remains a limitation when undertaking opioid-based pain therapies. Heroin, the diacetylated morphine derivative, has even stronger addictive properties and, as a major illicit drug of abuse, represents a public health problem worldwide.

Opiate drugs hijack a complex neuromodulatory system composed of three receptors, mu, delta, and kappa, which interact with a family of endogenous opioid peptides known as β-endorphin, enkephalins, and dynorphins. Opioid receptors form a subfamily of G-protein-coupled receptors (GPCRs) that also includes the homologous but non-opioid nociceptin/orphanin FQ receptor. Both receptors and peptides are expressed throughout peripheral and central nervous systems [1] and have been the subject of intense investigations for several decades. Opioids play a central role in pain processing and regulate many other aspects of physiology that include stress responses, respiration, gastrointestinal transit, and endocrine and immune functions [2]. The mood-regulating properties of endogenous opioids represent another main aspect of opioid physiology. The potent euphoric effects of known opiate drugs, and the high density of peptides and receptors in limbic brain areas, set the opioid system as a central player in both reward processing and mood control (see Glossary), and a feasible target for treatment of emotional dysfunction [3].

Mood disorders are defined as a group of diagnoses in which mood disturbance is the main underlying feature. Among these, major depressive disorder (MDD) is a chronic relapsing disorder characterized by the repetition of major depressive episodes (MDEs). The natural course of a depressive episode is remission (50% in 1 year), even in the absence of medical intervention. Unfortunately, 80% of patients experience relapse within 15 years [4]. Depression is a worldwide leading cause of disability, expected to even worsen in the next decades [5]. In the early 1900s, an opioid cure was proposed for the treatment of depressed patients through progressive exposure to low doses of an opiate mixture. Although seemingly effective [6], this approach was hampered by the inherent addictive properties of available opiates, as is the case for pain treatment. Since the advent of monoamine-targeting drugs in the 1950s, the antidepressant utility of opiates has been considered less, and first-line treatments in modern medicine currently rely on selective serotonin reuptake inhibitors (SSRIs). However, SSRIs are effective in only 40–50% patients [7] and identification of new targets for therapeutic intervention remains a major challenge.

Today, opiates are re-entering the therapeutic arsenal for MDD treatment. Drugs such as buprenorphine are used in the specific contexts of refractory depression [8] and depression–addiction comorbidity [9], and may have broader indications. Recent clinical and animal research data further strengthen the notion that endogenous opioids contribute to the etiology of mood disorders. Here we review both genetic and pharmacological approaches that reveal mu, delta and kappa opioid receptors as highly distinct players in reward processes and emotional responses, and very different targets for clinical intervention in MDD. The role of endogenous opioids has been partly reviewed elsewhere [10] and is only briefly mentioned when appropriate.

Section snippets

Opioid receptor knockout studies in behavioral models of reward and mood disorders

Constitutive knockout (KO) mice for MOR, DOR, and KOR were created almost two decades ago [11] and have been characterized for reward processes and mood states 1, 12, 13, which both involve hedonic responses (Figure 1) [14].

Data from MOR KO mice concur to establish that this receptor represents a key molecular player for reward processing of both natural stimuli and drugs of abuse. This robust activity contributes to recreational drug use and facilitates the onset of addictive behaviors [15].

Antidepressant potential for MOR, DOR, and KOR drugs

Pharmacological data (Table 1) demonstrate the effects of opioid drugs in rodent models of depression. Historically, antidepressant-like effects of enkephalins and endorphins were first reported for FS in rats [29]. Inhibitors of enkephalinase [30] and the general opioid antagonist naloxone [31] were later found to have antidepressant- and depressant-like effects, respectively, in the learned helplessness (LH) model. These results indicated that endogenous opioid peptides regulate despair-like

Opioid receptors and the neurobiology of depression

Many neurotransmitter systems and brain sites are involved in the pathogenesis of depression [58] and opioid receptors regulate a number of these aspects (Figure 2a,b). Relevant to depression, opioid receptors also regulate activity of the hypothalamus–pituitary–adrenal gland (HPA) axis, a major endocrine stress system. This aspect of opioid receptor function, as well as implications for mood, has been reviewed elsewhere 27, 59.

Addiction–depression comorbidity

Comorbidity is extremely frequent in psychiatry and the association between depression and addiction is one of the most classical clusters [131]. According to the self-medication hypothesis, depressed patients may take advantage of acute euphoric properties of drugs of abuse to overcome their depressed mood, a transient relief that bears its own risk of entering into dependence [25]. Conversely, repeated exposure to drugs of abuse triggers neuroadaptations in brain structures that contribute to

Concluding remarks

As behavioral models in rodents become more sophisticated and neurobiological mechanisms of mood disorders are better understood, MOR, DOR, and KOR appear as important and highly distinct players in the regulation of emotional states. In brief, animal studies show that DOR improves mood states acutely, KOR decreases mood after stress, and MOR exerts contrasting effects on mood. Clinical trials are needed to evaluate potential benefits of DOR agonist and KOR antagonist strategies to treat

Acknowledgments

We would like to thank Sandra Bour for her help with figure preparation and Claire Gaveriaux-Ruff for critical reading of the manuscript. This work was supported by CNRS, INSERM, and Université de Strasbourg. We also thank the Mouse Clinical Institute (ICS, Illkirch, France), the European Union (Grant No. GENADDICT/FP6 005166), Fondation pour la Recherche Médicale (P.E.L.), Fondation Fyssen (P.E.L.), and the National Institutes of Health (National Institute of Drug Addiction, grant #05010 and

Glossary

Anhedonia
reward-related deficit characterized by markedly diminished interest or pleasure in all, or almost all, activities.
Biased agonism
well-recognized phenomenon whereby GPCRs exist under distinct agonist-dependent active conformations that form specific agonist–receptor–effector complexes that in turn induce distinct and agonist-specific downstream signaling and regulatory responses.
Chronic mild stress (CMS)
animals are subjected to a variety of unpredictable physical and social stressors,

References (153)

  • A. Markou

    Neurobiological similarities in depression and drug dependence: a self-medication hypothesis

    Neuropsychopharmacology

    (1998)
  • A.T. Knoll et al.

    Dynorphin, stress, and depression

    Brain Res.

    (2010)
  • M.R. Bruchas

    The dynorphin/kappa opioid system as a modulator of stress-induced and pro-addictive behaviors

    Brain Res.

    (2010)
  • A.J. Kastin

    Enkephalin and other peptides reduce passiveness

    Pharmacol. Biochem. Behav.

    (1978)
  • P. Tejedor-Real

    Effect of mixed (RB 38A) and selective (RB 38B) inhibitors of enkephalin degrading enzymes on a model of depression in the rat

    Biol. Psychiatry

    (1993)
  • P. Tejedor-Real

    Implication of endogenous opioid system in the learned helplessness model of depression

    Pharmacol. Biochem. Behav.

    (1995)
  • D.C. Broom

    Nonpeptidic delta-opioid receptor agonists reduce immobility in the forced swim assay in rats

    Neuropsychopharmacology

    (2002)
  • E.M. Jutkiewicz

    Behavioral and neurobiological effects of the enkephalinase inhibitor RB101 relative to its antidepressant effects

    Eur. J. Pharmacol.

    (2006)
  • P.S. Naidu

    NIH 11082 produces anti-depressant-like activity in the mouse tail-suspension test through a delta-opioid receptor mechanism of action

    Eur. J. Pharmacol.

    (2007)
  • M.M. Nieto

    Physiological control of emotion-related behaviors by endogenous enkephalins involves essentially the delta opioid receptors

    Neuroscience

    (2005)
  • A. Saitoh

    Potential anxiolytic and antidepressant-like activities of SNC80, a selective delta-opioid agonist, in behavioral models in rodents

    J. Pharmacol. Sci.

    (2004)
  • A. Saitoh

    The novel delta opioid receptor agonist KNT-127 produces antidepressant-like and antinociceptive effects in mice without producing convulsions

    Behav. Brain Res.

    (2011)
  • A. Saitoh

    Antidepressant-like effects of the delta-opioid receptor agonist SNC80 ([(+)-4-[(αR)-α-[(2S, 5R)-2,5-dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N,N-diethylbenzamide) in an olfactory bulbectomized rat model

    Brain Res.

    (2008)
  • P. Tejedor-Real

    Involvement of delta-opioid receptors in the effects induced by endogenous enkephalins on learned helplessness model

    Eur. J. Pharmacol.

    (1998)
  • M.M. Torregrossa

    Peptidic delta opioid receptor agonists produce antidepressant-like effects in the forced swim test and regulate BDNF mRNA expression in rats

    Brain Res.

    (2006)
  • R. Vergura

    Anxiolytic- and antidepressant-like activities of H-Dmt-Tic-NH-CH(CH2-COOH)-Bid (UFP-512), a novel selective delta opioid receptor agonist

    Peptides

    (2008)
  • Q.Z. Yang

    The antidepressant-like effect of human opiorphin via opioid-dependent pathways in mice

    Neurosci. Lett.

    (2011)
  • M.O. Rojas-Corrales

    Antidepressant-like effects of tramadol and other central analgesics with activity on monoamines reuptake, in helpless rats

    Life Sci.

    (2002)
  • S.C. Ribeiro

    Interface of physical and emotional stress regulation through the endogenous opioid system and mu-opioid receptors

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2005)
  • E.J. Nestler et al.

    The mesolimbic dopamine reward circuit in depression

    Biol. Psychiatry

    (2006)
  • L. Cordonnier

    Facilitation of enkephalins-induced delta-opioid behavioral responses by chronic amisulpride treatment

    Neuroscience

    (2005)
  • L. Hipolito

    Shell/core differences in mu- and delta-opioid receptor modulation of dopamine efflux in nucleus accumbens

    Neuropharmacology

    (2008)
  • R. Tao et al.

    Involvement of the dorsal raphe but not median raphe nucleus in morphine-induced increases in serotonin release in the rat forebrain

    Neuroscience

    (1995)
  • K. Grasing et al.

    Selegiline prevents long-term changes in dopamine efflux and stress immobility during the second and third weeks of abstinence following opiate withdrawal

    Neuropharmacology

    (1998)
  • S.R. Hodgson

    Different affective response to opioid withdrawal in adolescent and adult mice

    Life Sci.

    (2009)
  • C. Goeldner

    Impaired emotional-like behavior and serotonergic function during protracted abstinence from chronic morphine

    Biol. Psychiatry

    (2011)
  • P.E. Lutz

    Sequential and opposing alterations of 5-HT1A receptor function during withdrawal from chronic morphine

    Eur. Neuropsychopharmacol.

    (2011)
  • M.R. Bruchas

    Selective p38α MAPK deletion in serotonergic neurons produces stress resilience in models of depression and addiction

    Neuron

    (2011)
  • G.C. Harris et al.

    Activation in extended amygdala corresponds to altered hedonic processing during protracted morphine withdrawal

    Behav. Brain Res.

    (2007)
  • G.C. Harburg

    Knockout of the mu opioid receptor enhances the survival of adult-generated hippocampal granule cell neurons

    Neuroscience

    (2007)
  • T.P. Cominski

    Loss of the mu opioid receptor on different genetic backgrounds leads to increased bromodeoxyuridine labeling in the dentate gyrus only after repeated injection

    Neuroscience

    (2012)
  • A. Dranovsky

    Experience dictates stem cell fate in the adult hippocampus

    Neuron

    (2011)
  • P. Song et al.

    The involvement of glial cells in the development of morphine tolerance

    Neurosci. Res.

    (2001)
  • H. Zhang

    Central kappa-opioid receptor-mediated antidepressant-like effects of nor-binaltorphimine: behavioral and BDNF mRNA expression studies

    Eur. J. Pharmacol.

    (2007)
  • M.S. Ansorge

    Neurodevelopmental origins of depressive disorders

    Curr. Opin. Pharmacol.

    (2007)
  • T.L. Bale

    Early life programming and neurodevelopmental disorders

    Biol. Psychiatry

    (2010)
  • B. Klausz

    Changes in adaptability following perinatal morphine exposure in juvenile and adult rats

    Eur. J. Pharmacol.

    (2011)
  • J. Le Merrer

    Reward processing by the opioid system in the brain

    Physiol. Rev.

    (2009)
  • E. Berrocoso

    Opiates as antidepressants

    Curr. Pharm. Des.

    (2009)
  • T.I. Mueller

    Recurrence after recovery from major depressive disorder during 15 years of observational follow-up

    Am. J. Psychiatry

    (1999)
  • Cited by (381)

    View all citing articles on Scopus
    View full text