You are on page 1of 16

Reviews

Opioid receptors: drivers to addiction?


Emmanuel Darcq1 and Brigitte Lina Kieffer   1,2*
Abstract | Drug addiction is a worldwide societal problem and public health burden, and results
from recreational drug use that develops into a complex brain disorder. The opioid system, one of
the first discovered neuropeptide systems in the history of neuroscience, is central to addiction.
Recently , opioid receptors have been propelled back on stage by the rising opioid epidemics,
revolutions in G protein-​coupled receptor research and fascinating developments in basic
neuroscience. This Review discusses rapidly advancing research into the role of opioid receptors
in addiction, and addresses the key questions of whether we can kill pain without addiction using
mu-​opioid-receptor-​targeting opiates, how mu- and kappa-​opioid receptors operate within
the neurocircuitry of addiction and whether we can bridge human and animal opioid research
in the field of drug abuse.

Psychotomimetic
Opium is extracted from the seeds of poppies (Papaver Distributed throughout the nervous system, opioid pep-
An effect caused by drugs, somniferum) and has been used for more than 4,000 years tides and receptors reduce responses to painful stimuli
mimicking symptoms of in medicinal and recreational practices to relieve pain and stress, and influence reward processing and mood.
psychosis, such as agitation, and cause euphoria. Morphine was isolated in 1805 as the The latter influence represents a fundamental role of the
delusions and delirium.
most active component of opium1 and remains the most opioid system and will be a main focus of this Review.
potent painkiller in modern medicine, despite severe Note that the activity of the endogenous opioid system is
adverse effects and a strong potential for addiction. extremely broad and covers many other aspects of phys-
Heroin, the diacetylated form of morphine, was origi- iology and behaviour (reviewed in ref.9), but these are
nally marketed as the first non-​addictive opiate to treat less related to addiction and will not be discussed here.
cough and asthma in 1898, yet heroin addiction has In the late 1980s and early 1990s, the isolation
represented a major societal problem ever since. of three genes encoding opioid peptide precursors
More recently, an ‘opioid epidemic’ has emerged in (namely, POMC, PENK and PDYN, which encode
occidental countries, particularly in North America2. proopio­melanocortin, preproenkephalin (also known as
The overprescription of opioids for pain relief in the proenkephalin) and preprodynorphin (also known
past 20 years has led to a rapid surge in the non-​medical as prodynorphin), respectively) and the genes encoding
use of prescribed opioids, with deaths by overdose and MORs (OPRM1), DORs (OPRD1) and KORs (OPRK1)
transition to heroin abuse rising at alarming rates3–6. opened an era of molecular and genetic investigations
The increasing availability of low-​cost synthetic opioids, of the opioid system10,11. Oprm1 deletion in mice simul-
such as non-​pharmaceutical fentanyls7, further feeds the taneously eliminated the analgesic, rewarding and
epidemic. This opioid crisis has fostered novel public dependence-​inducing effects of morphine12, demon-
policies and much interest in developing better opioids strating that the MOR is the sole responsible receptor
to treat pain. For medical purposes8, the ideal opiate for both the therapeutic and the adverse actions of
drug would relieve pain with high and sustained efficacy morphine. The MOR is also the key molecular target
1
Douglas Mental Health (that is, without tolerance), without the threats of res- for biological effects of other clinically useful and/or
Institute, Department of piratory depression (the main cause of overdose) and abused opiates (including heroin, fentanyl, oxycodone
Psychiatry, McGill University,
Montreal, Quebec, Canada.
without drug dependence (contributing to addiction). and methadone). Given the high risk of adverse effects of
The early 1970s saw the game-​changing discovery MOR agonists, the enthusiasm for drug discovery efforts
2
Institut de Génétique et de
Biologie Moléculaire et that opiate drugs bind to receptors in the brain (see ref- targeting MORs abated in the late 1990s (although new
Cellulaire, INSERM, Centre erences cited in ref.1) and hijack a complex endogenous efforts are starting; see Box 1).
National de la Recherche neuromodulatory system. The opioid system comprises On another front, Oprd1-knockout mice revealed that
Scientifique and University of three homologous G protein-​coupled receptors (GPCRs) DORs have anxiolytic and antidepressant functions13,
Strasbourg, Strasbourg,
France.
known as mu-, delta- and kappa-​opioid receptors decidedly distinguishing this receptor from MORs. Many
(MORs, DORs and KORs, respectively). Under physio­ pharmacological studies have now implicated DORs in
*e-​mail: brigitte.kieffer@
douglas.mcgill.ca logical conditions, opioid receptors are stimulated by mood disorders and chronic pain14–16. By contrast, KOR
https://doi.org/10.1038/ endogenous opioid peptides, forming a peptide family activation produces both aversive and psychotomimetic
s41583-018-0028-x that includes β-​endorphin, enkephalins and dynorphins. effects17. This peculiar profile has strongly limited the

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 499


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | Can we kill pain without addiction using MOR opiates?


Many drug discovery programmes targeting mu-​opioid receptors (MORs) Other approaches
for analgesia without addiction have had limited success. However, recent Other approaches163 (part c of the figure) include identifying
advances in G protein-​coupled receptor (GPCR) biology have revitalized compounds acting at multiple opioid receptors, opioid receptor dimers
the field. (MOR–delta-opioid receptor (DOR); see Supplementary Box 1) or
opioid–non-opioid receptor dimers (for example, MOR–CC-​chemokine
Biased signalling
receptor 5 (CCR5)); compounds with agonist and antagonist activity at
GPCR conformation depends on biological context. The active complex is
MORs and ‘anti-​opioid’ receptors (such as cholecystokinin, neurokinin 1
determined by the drug and neighbouring proteins, including effectors150.
or the nociceptin peptide receptor), respectively; and compounds
Thus, different agonists may engage distinct effectors, leading to
targeting effectors besides arrestins (for example, regulator of G
biased agonism (see part a of the figure). Biased MOR agonists might
protein signalling (RGS) proteins). In addition, a compound binding a
reduce pain with minimal adverse effects151–153 (part b of the figure). Mice
truncated MOR isoform (lacking the first transmembrane domain) had
lacking β-​arrestin 2 (β-​arr) — one of the two main MOR effectors —
an encouraging pharmacological profile, although with unusual
showed enhanced morphine analgesia154 and reduced morphine-​induced
properties164,165; the biological importance of this isoform remains
constipation and respiratory depression155. Gi-​biased MOR agonists that
unclear166.
minimally recruit β-​arr include TRV130 (also known as oliceridine;
In a novel approach (part c of the figure), the active MOR receptor
identified through cell-​based assays)156, which is in a phase III trial for
structure167 was used to computationally simulate ligand docking at low
analgesia157, mitragynine pseudoindoxyl (developed from natural
pH, with the rationale that binding in an acidic environment might limit
products)158 and PZM21 (identified using computational docking)26. These
effects to injured sites. A pH-​sensitive fluorinated fentanyl derivative
compounds confer strong analgesia with reduced constipation and
called NFEPP reduced inflammatory pain without central or intestinal
respiratory depression in rodents. Novel MOR agonists covering the entire
effects168.
Gi–β-​arr bias range were recently designed, with Gi bias correlating with
Adverse effects might be limited by mimicking or enhancing
the analgesia–respiratory depression therapeutic window159.
endogenous opioid signalling (part d of the figure). Endogenous opioid
Biased signalling in cells has not often been correlated with
analogues with improved stability and bioavailability have been
addiction-related behaviours (for example, drug reward), as these in vivo
developed163. Recently, MOR positive allosteric modulators were
experiments are less amenable to medicinal chemistry efforts. The three
developed to act on allosteric sites (2) to facilitate agonism at the
compounds in part b do not induce conditioned place preference at
orthosteric site (1), strengthening MOR activity at the optimal times
analgesic doses26,158. However, further research should assess the hedonic
and sites, with fewer adverse effects (part d of the figure)169.
and motivational properties of Gi-​biased drugs; a first study has mixed
BMS-986122 enhances opioid peptide-​induced signalling170 and
conclusions160. Moreover, part of the gap between in vitro predictions and
probably binds to the MOR Na+-binding site171. In vivo studies will
in vivo responses depends on location bias161 and systems bias162. Studies
show whether the concept holds to develop safer opioid analgesics.
on receptor signalling in different neuronal compartments and at different
Part a is adapted with permission from ref.172, Elsevier.
brain sites will expand in the future.

a Biased signalling and drug design c Other approaches for better MOR analgesics
Drug 1 Drug 2 Drug 3 • Multifunctional opioids Clinical and
Gi/o-biased opioids
• Dimer-specific opioids abused opioids
R R
R
E2 E3 E2 Opioid
E1 E1
specific to
‘acidic’ receptor
R MOR
Low efficacy Optimal Adverse effects

b Biased signalling at MOR


Signalling regulator
HO Anti-opioid system
N
CH3
O H O Behaviour
O NH
N CH
HO
3
S d Endogenous signalling and allostery at MOR
Morphine Oliceridine Endogenous opioid
Endogenous
O CH3 S
opioid analogue
MOR N N
N H H
HO H3C CH3 2
1
PZM21
β-Arr Gi/o Positive allosteric
CH3 modulator
O
N
S
• Constipation H3 C O N
NH O CH3 O
• Respiratory S
depression Analgesia O Br
O
H3C O Temporally and spatially O CH3
• Reward? Mitragynine pseudoindoxyl controlled enhanced Cl
BMS-986122
• Tolerance and/or dependence? signalling
E, effector; R, receptor.

500 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Biased agonism development of KOR agonists for pain control and has that drive decision making. Aminergic neurotrans-
A signalling response given KORs a reputation of being associated with the mitter systems (such as the dopamine (DA) and
determined by the ‘dark side’ of emotional and perceptual experience18,19 serotonin (5-HT) systems) have been extensively
conformation of the drug– (Box 2). Overall, the very distinct profiles of MORs, DORs studied in this context. Circuit mechanisms encod-
receptor–effector complex that
engages only a subset of
and KORs have now been clarified, and opportunities in ing reward and/or aversion involve the activity and
cellular effectors. Some opioid research are rapidly advancing. interaction (sometimes bidirectional) of overlapping
high-throughput screening The past decade has witnessed revolutions in GPCR networks32. The opioid system interacts anatomically
programmes have aimed to structure and signalling research20,21. In 2012, the first and functionally with reward–aversion networks33,34,
design novel ‘biased’ drugs with
atomic structures of the MOR22, DOR23, KOR24 and the to maintain hedonic balance, regulate mood states and
improved therapeutic profiles.
structurally related nociceptin/orphanin FQ receptor25 cope with stress (Fig. 2a). Furthermore, the notions
Therapeutic window (Box 3) solved by X-​ray crystallography were published that, for example, endogenous opioids contribute to
Dose range for a drug that (Fig. 1a), opening the way to design opioid drugs with the rewarding effects of pain relief35 and that phys-
allows therapeutic efficacy with entirely novel chemical scaffolds (for example, see ref.26) ical and social pain (such as that associated with
no (or minimal) side effects.
and perhaps to get closer to the ideal analgesic (Box 1). social rejection) share common opioid-​m ediated
Location bias Many signalling effectors activated by opioid receptor mechanisms36,37 are gaining interest.
Bias in receptor signalling stimulation have been identified (for example, those Overall, the opioid system can be considered a
dictated by the location of the in Fig. 1b). Furthermore, new technologies (such as central regulator in basic processes of individual and
receptor in the cell (for example,
neural tracing, connectome analysis, optogenetics and species survival, geared to enhance reward-​b ased
at the surface or in endosomes
or Golgi) and the availability of
chemogenetics) enabled the dissection of neural circuit learning and reduce aversive experiences. Opioid
effectors at this site. organization and function at microscale or macroscale receptors are therefore critical in the emergence of
levels27–29, and the notion of precision medicine is gradually neuropsychiatric disorders that manifest when reward
Systems bias entering the areas of pain30 and addiction31 treatment. and aversion processing are dysfunctional. These
Bias in receptor signalling
driven by anatomical
Opioid research strongly benefits from advances in these pathologies primarily include addiction and depres-
localization within brain circuits domains, and this Review addresses three questions that sion, and opioid receptor targets have therapeutic
subserving the behavioural arguably foster the strongest interest in current opioid potential in both of these disease areas. Opioid recep-
response and the effectors research. First, can we kill pain without addiction using tor function in mood disorders has been reviewed
available at those sites.
MOR-​targeting opiates (addressed in Box 1)? Second, elsewhere38.
Precision medicine how do MORs and KORs operate within the neuro­
Also known as personalized circuitry of addiction? Third, can we bridge human and Opioid receptors and the multiple faces of addiction.
medicine. An innovative animal opioid research in the field of drug abuse? Addiction is a complex, relapsing disorder in which
approach in medicine in which drugs of abuse hijack, overstimulate and compromise
interindividual variability (in
lifestyle, environment and
Opioid receptors in addiction circuits reward-​processing systems and associated networks.
genes) is taken into Experimental evidence from animal research has posi- The disease develops from an initiation phase, during
consideration for disease tioned the role of the opioid system at the centre of reward which the drug produces pleasurable effects and is
prevention and/or treatment. and aversion processing, and has highlighted the notion consumed recreationally. Upon repeated consumption,
that the dysregulation of opioid neurotransmission is a control over drug taking is gradually lost, leading to
Hedonic balance
The equilibrium between
main driver to drug abuse (Fig. 2). compulsive drug seeking and drug taking39,40 (Fig. 2b).
positive and negative affect. Whereas recreational drug use is essentially motivated
A positive hedonic state is Opioid system and reward–aversion in physiology. by reward seeking, drug intake in individuals who are
considered a state of well-​ Searching for reward (such as food, sex and social addicted is also driven by other factors that arise owing
being, whereas a negative
hedonic state is unpleasant.
interactions) and avoiding punishment or discom- to brain adaptations to chronic drug exposure (Fig. 2b).
fort (for example, pain) are two fundamental forces These include reduced self-​control41, enhanced incen-
tive salience (that is, importance of the context) and
habit formation42, altered reward processing and stress
Box 2 | The intriguing hallucinogenic properties of KOR agonists reactivity43, and the emergence of a negative affective
Beyond aversion, kappa-​opioid receptor (KOR) agonists show hallucinogenic properties state upon withdrawal19 or with protracted abstinence
— a facet of KOR function that is unique among opioid receptors. Salvinorin A is a (see ref.44 and references therein).
natural product from the sage Salvia divinorum, or ‘magic mint’, that was long used by Together, alterations of both positive and neg-
the Mazatecs of Oaxaca, Mexico for spiritual rituals and medicinal practice, and was ative affect contribute to the development and
recently discovered to be a specific KOR agonist173,174. Salvinorin A is a highly potent maintenance of addiction, and relevant transmitter
hallucinogen for which the psychoactive effect is comparable to that of lysergic acid
and circuit adaptations are being actively studied 39.
diethylamide (LSD). The use of salvinorin A, which is much less regulated than, for
All three opioid receptors are involved in the process,
example, LSD use, is currently gaining popularity among young individuals in a
recreational context. although with very different contributions (Fig. 2b).
The finding that this KOR agonist has psychoactive effects definitively establishes a The effects of MORs and KORs in regulating addic-
specific role for KORs in higher cognitive and perceptual functions, the dysregulation tion networks and, more importantly, the neuronal
of which is associated with psychiatric disorders such as psychosis, bipolar disorders populations in which these receptors operate, have
and dementia175. This particular KOR function remains poorly understood and adds been well characterized and are discussed below
another aspect to the known role of this receptor in stress responses, mood deficits and (Fig. 2c). DOR-​mediated circuit mechanisms contrib-
drug abuse. The circuits underlying this phenomenon are yet to be characterized. uting to addiction have been less well studied using
Notably, new analogues of salvinorin A with partial agonism or biased ligand properties cell-​specific genetic approaches (only one study 45);
are now proposed for the treatment of neuropsychiatric diseases, including
thus, the current knowledge of DOR involvement in
addiction176; this research is still at an early stage.
addiction is reviewed more briefly.

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 501


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Reverse pharmacology
MORs in reward, motivation and self-​control. MORs naloxone aversion in a conditioned place aversion (CPA)
An approach in which a mediate the pleasurable properties of therapeutic and/or paradigm, indicating that MORs are crucial in mediat-
receptor or endogenous ligand abused opiates in vivo. Importantly, genetic approaches ing the positive hedonic tone elicited by endogenous
is discovered first, the have demonstrated that these receptors are also necessary opioids52. Together, the findings above show that MOR
physiological function is
for the rewarding effects of other drugs of abuse activity both mediates natural rewards and promotes
determined.
(reviewed elsewhere11,46 and in Supplementary Table 1). recreational drug use, with the latter in turn favouring
Conditioned place Briefly, Oprm1-knockout mice show lower levels of vol- the onset of drug abuse.
preference untary alcohol drinking and self-​administration (SA) MORs are expressed throughout the addiction cir-
(CPP). A behavioural paradigm
and fail to express conditioned place preference (CPP) in cuitry53 (Fig. 2c). Several brain sites for MOR-​mediated
in rodents that determines the
rewarding or aversive effect of
response to tetrahydrocannabinol (THC) or nicotine. The reward have been identified by local pharmacology, and
a drug on the basis of time latter molecules primarily activate non-​opioid receptor these mainly belong to mesocorticolimbic networks54.
spent in a drug-​associated targets (that is, cannabinoid receptors and nicotinic Central to this circuitry is the well-​characterized ven-
context after conditioning. receptors, respectively), and these activated receptors, tral tegmental area (VTA)–nucleus accumbens (NAc;
in turn, trigger opioid release at appropriate MOR-​ also known as the ventral striatum) DA pathway. The
Tetrahydrocannabinol
(THC). The principal expressing brain sites to produce reward11. The data prevailing disinhibition hypothesis postulates that
psychoactive component of are less clear for cocaine and amphetamine, as Oprm1- activation of MORs expressed by VTA GABAergic
cannabis, which produces knockout mice show decreased SA for these substances, interneurons relieves the local inhibitory tone and
central effects by acting at
but no change in CPP (see ref.11), consistent with the thus disinhibits DA neurons, which release DA to sig-
cannabinoid CB1 receptors.
notion that psychostimulants hijack reward systems via nal drug reward55,56. This hypothesis is supported by
Psychostimulants mechanisms that do not necessarily engage MORs, at many pharmacology and electrophysiology studies57.
A group of substances least for their rewarding effects47,48. However, this is not the only mechanism through which
(including cocaine and Genetic approaches have also demonstrated the MORs affect DA signalling, as MORs are also abundant
amphetamines) that enhance
essential role of MORs in mediating natural rewards. in brain areas that receive DA neuron projections and,
physical and cognitive
performance. Psychostimulants The constitutive Oprm1 deletion reduced maternal notably, among NAc neurons that project back to the
are used to treat attention attachment in 4–8 day old mutant pups49; furthermore, VTA58,59. Furthermore, DA-​independent opioid reward
deficit–hyperactivity disorder. adult knockout mice showed impaired social interac- is also documented60–63, but in fact little is known about
tions together with several other signs of autistic-​like MOR-​expressing cells driving opioid reward outside
behaviours50, confirming the essential function of MORs the VTA.
in social bonding that was previously suggested by the A few studies using genetic approaches in mice have
pharmacology36,38. Oprm1-null mutants also showed recently interrogated MOR function outside the VTA.
reduced motivation for both food and sucrose in an SA In the striatum, MOR expression is robust in DA D1
paradigm51. Finally, Oprm1-null mutant mice showed no receptor (D 1R)-expressing medium spiny neurons

Box 3 | The opioid system’s cousin: the opioid-​like receptor and nociceptin/orphanin FQ
The cloning of the opioid-​receptor-encoding genes led to the identification agonists reduce drug reward and basal or drug-​induced (mostly cocaine-​
of homologous G protein-​coupled receptor (GPCR)-encoding genes, induced) dopamine release in the nucleus accumbens, alleviate signs of
some of which were known (such as those encoding somatostatin alcohol withdrawal and diminish stress-​primed or drug-​primed
receptors). However, the closest homologue encoded a GPCR for which the reinstatement of cocaine place preference, possibly by antagonizing
ligand was unknown, thereafter named opioid-​like receptor (ORL1). This corticotropin releasing factor (CRF) stress systems. Furthermore, the fact
receptor does not bind opioids with high affinity, and therefore cannot be that N/OFQ blocks morphine-​induced supraspinal analgesia and
classified as an opioid receptor. Using reverse pharmacology177, two teams conditioned place preference suggests that the N/OFQ–NOP system acts
discovered its endogenous ligand, a 17-amino-​acid peptide named either as an anti-​opioid system for some responses. Indeed, NOP genetic
orphanin FQ178 or nociceptin179 based on its first reported characteristics inactivation or blockade reduces analgesic tolerance to morphine,
(its peptide sequence and its effect on pain). This was one of the first GPCR confirming functional interactions between the two systems. Rodent
de-​orphanizations, and the receptor and peptide are now classically known research has therefore positioned the NOP system as a feasible target for
as the nociceptin opioid peptide receptor (NOP) and N/OFQ, respectively. the development of addiction treatments.
Strikingly, just as NOP shows the highest homology with the kappa-​ At this stage, however, translation to non-​human primates and the clinic
opioid receptor (KOR), the N/OFQ peptide shows sequence similarity may prove challenging for two reasons. First, the anatomical localization
with dynorphin (although lacking the amino-​terminal tyrosine typical of of the N/OFQ–NOP system seems to be different in rodent, non-​human
opioid peptides), suggesting a common ancestor in evolution. Similar to primate and human brains. The recent synthesis of positron emission
the opioid receptors, NOP is an inhibitory Gi/o-​coupled GPCR that reduces tomography (PET) tracers for NOP will help to guide clinical trials180.
neuronal activity and/or neurotransmitter release. Many synthetic NOP Second, the effects of N/OFQ on the drug-​abuse-associated aspects of
ligands developed in academia and industry180, as well as genetic mouse physiology (such as stress and anxiety) have proved to be complex; both
mutants, have been used to study the multiple in vivo functions of the NOP agonists and antagonists need be examined for their potential
system and its potential interactions with the opioid system (reviewed in therapeutic utility. The current thinking is that NOP blockade has
ref.181). NOP-​regulated physiology and pathology cover areas of pain potential for treating depression and obesity, whereas NOP activation
control, reward processing and drug abuse, stress, anxiety and mood may reduce anxiety and help to treat several aspects of addiction,
disorders, feeding and obesity, motor control and cognition, and operate including a reduction of consumption and the prevention of relapse180,182;
broadly in the nervous system (reviewed in refs181,182). To the best of our however, the best valid strategy remains open. As an illustration, a NOP
knowledge, no cell-​specific genetic studies have yet addressed NOP agonist (cebranopadol; phase III) and a NOP antagonist (JTC-801; phase II)
function in defined brain networks or neuronal populations. have both reached clinical trials to treat pain, and the NOP antagonist
Much evidence supports a role for the N/OFQ–NOP system in addiction LY2940094 was tested in phase II trials for major depressive disorders and
and this has been reviewed extensively180–182. In brief, N/OFQ and NOP for alcohol dependence180.

502 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

(so-​called D1 MSNs) and is barely detectable in DA partially restores remifentanil SA58, suggesting that
D2 receptor (D2R)-expressing MSNs (D2 MSNs)58,59. MORs expressed in the striatonigral pathway (that is,
Rescuing MOR expression in D1 MSNs of Oprm1- by D1 MSNs) are sufficient to mediate opioid reward.
knockout mice using a bacterial artificial chromosome However, whether this receptor population is neces-
Pdyn-​MOR transgene restores morphine CPP and sary for drug reward is less clear. Conditional Oprm1

a First solved atomic structure of opioid receptors


MOR–β-FNA DOR–naltrindole KOR–JDTic NOP–C-24
ECL2 ECL2
ECL3 1 ECL2
ECL1 ECL1
7 7 ECL3
EC membrane C-24
1 S–S

1
3
2 5
2 7
3 6 4
2 NPXXY
3
8 DRY ICL3
Helix 8 6
5 ICL1 IC membrane
5 ICL2
6

b The active MOR and signalling effectors


Ca2+ K+ MOR
ECL2 channel channel

AC
Gαi/o Arr GRK
1 G protein- ERK
7 dependent
5 JNK
signalling
6 G protein-
independent
signalling

CAMKII STAT3 PKA PKC p38 MAPK


10 Å

Fig. 1 | Opioid receptor structure and signalling. a | First resolved atomic All three opioid receptors are inhibitory-​type GPCRs, the activation of which
structures of inactive opioid receptors and the nociceptin opioid peptide reduces postsynaptic neuronal excitability or presynaptic neurotransmitter
receptor (NOP); see Box 3 ). The receptors are bound to their specific release151. At the cellular level, opioid receptors activate G protein-​
antagonists — β-​funaltrexamine (β-​FNA) for the mu-​opioid receptor dependent pathways involving both Gαi/o and Gβγ subunits, as well as G
(MOR)22, naltrindole for the delta-​opioid receptor (DOR)23 and JDTic for the protein-​independent signalling cascades that involve scaffold proteins such
kappa-​opioid receptor (KOR)24 as well as the peptide mimetic antagonist as arrestins (Arr). Altogether, many downstream signalling effectors have
Compound 24 (C-24) for the NOP receptor25. The receptors each have seven been identified for each receptor8,72,184,185. The scheme illustrates currently
transmembrane domains (numbered). DRY and NPXXY, shown here for the known effectors for MOR and indicates which are G protein-​dependent
KOR structure, are conserved motifs important for receptor function. (green) or G protein-​independent (red) or whether G protein and/or
b | MOR activation and signalling in cells. The left side of the panel shows a Arr dependency is unclear (orange). The cell adapts to repeated
comparison of the seven-​helical arrangement of active (green) and inactive receptor stimulation, leading to desensitization of receptor signalling
(blue) forms of the receptor167. The main modification is a 10 Å outward shift (via, for example, receptor trafficking and effector uncoupling) and/or
of the intracellular part of transmembrane domain 6, which typically compensatory upregulation of related cellular pathways72. As for most
interacts with cellular effectors and notably the Gα subunit of G proteins. GPCRs, opioid receptor activation is subjected to biased agonism; that is,
The conformation of the extracellular receptor domains shows minimal the cellular and in vivo responses are often agonist-​dependent185. AC,
changes. The structure also revealed a network of polar amino acid residues, adenylyl cyclase; CAMKII, calmodulin-​dependent protein kinase II; EC,
which links the binding site to the cytoplasmic face of the receptor and is extracellular ; ECL , extracellular loop; ERK , extracellular-​signal-regulated
involved in G protein-​coupled receptor (GPCR) signal propagation167. The kinases; GRK , GPCR kinase; IC, intracellular ; ICL , intracellular loop; JNK ,
switch from inactive to active structures was also probed using solution-​ Jun N-​terminal kinase; MAPK , mitogen-​activated protein kinase; PKA ,
state NMR , revealing that the conformational changes of transmembrane protein kinase A ; PKC, protein kinase C; STAT3, signal transducer and
domains 5 and 6, the main receptor movements to reach full activation, activator of transcription 3. Structures in part a adapted from refs22–25,
require engagement of both the ligand and the G protein mimic in the Macmillan Publishers Limited. Structure in part b adapted from ref.167,
complex183. The right side of the panel illustrates MOR signalling in cells. Macmillan Publishers Limited.

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 503


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

a Opioid system physiology b Opioid receptors in the disease process


Reward Aversion
MOR
Euphoria
• Adaptions to the drug
• Lower reward • Withdrawal • Increased dysphoria
Hedonic tone • Aversive state • Lower mood
Recreational • MOR adaption • Binge
KOR • Higher anxiety
• Dysphoria stress drug use MOR • Intoxication • Increased stress
• Negative affect reactivity
• Lower self-control
DOR Mood • Maladaptive habits KOR–Dyn
• Higher context salience • Preoccupation DOR
• Low anxiety
• Positive affect MOR/DOR/KOR? • Anticipation

c Opioid receptor function in addiction circuits

MOR HIP KOR HIP


FC HB FC HB

CP DRN CP DRN
NAc BNST NAc BNST
VTA VTA

AMY AMY

Preoccupation or anticipation Withdrawal or aversive state Receptor density


Binge or intoxication Low Medium High

Fig. 2 | Opioid receptors in physiology and addiction. a | Opioid receptors drug cue or context learning103. c | MOR and KOR function in neurocircuits
regulate reward and aversion. The opioid system contributes to self and of addiction. This simplified scheme summarizes animal data involving
species survival by promoting reward elicited by natural stimuli (such as food, genetic or cell-​specific approaches to study opioid receptors in circuits of
sex and social interaction), regulating mood states and facilitating efficient addiction-​related behaviours. Similar studies for the DOR are only just
coping with pain and stress. Mu-​opioid receptors (MORs) and kappa-​opioid starting45. Brain regions involved in drug abuse are represented (circles) with
receptors (KORs) oppositely regulate hedonic homeostasis, with MOR their known connectivities (light grey lines) and include most regions studied
agonists and KOR agonists producing euphoria and dysphoria, respectively. in rodent addiction research. Circles are outlined in different colours
Stress and drug abuse both enhance KOR–dynorphin (Dyn) signalling, depending on whether the brain structure has been associated with binge
contributing to an increase in dysphoric states43,83. By contrast, delta-​opioid or intoxication (orange), withdrawal or aversion (blue), or preoccupation or
receptor (DOR) activity reduces anxiety and depressive states, and regulates anticipation (red) stages and are based on information in refs39,40, with the
learning and memory14,105. KOR blockade and DOR activation therefore have addition of the habenula (HB) and dorsal raphe nucleus (DRN), which are
the potential to improve emotional responses. Finally , MORs and DORs show the focus of increasing interest in the context of aversive aspects of
contrasting activities on motor impulsivity (see main text). Opioid receptors addiction. Receptor density36,54 (represented in a naive mouse brain) is
are therefore prime targets for the treatment of addiction and disorders such indicated for each region, and the opioid-​receptor-regulated pathways
as depression38 that are characterized by low-​reward or high-​aversion states. identified in the studies discussed in this Review are shown by black arrows.
b | Opioid receptors in the addiction cycle. In highly simplified terms, In brief, MORs in GABAergic interneurons of the ventral tegmental area (VTA)
addiction can be characterized by ‘low-​reward, high-​aversion’ functioning, facilitate dopamine release and drug reward57, whereas MORs in the striatum
as negative affective states progressively overtake positive affect. A well-​ (nucleus accumbens (NAc) and caudate putamen (CP)), expressed mainly in
accepted view from animal research is that drug abuse unfolds as a three-​ dopamine D1-type neurons58,59, regulate both drug reward (for alcohol and
stage cycling process, involving binge or intoxication episodes, followed by morphine)58,64 and heroin seeking59. MOR adaptations in the DRN drive
withdrawal and a negative affective state when the drug clears, in turn depressive-​l ike states and social withdrawal in protracted heroin
leading to a preoccupation or craving step, during which desire for the drug abstinence74. The roles of MOR-​expressing cell populations in regions that
intensifies (craving) and triggers the next intoxication episode (inspired by modulate negative affective states, and in regions where the receptor is
refs39,40). Neuroplastic changes occur at all the stages, contributing to abundant (HB, DRN and amygdala (AMY)), remain to be studied. KORs in VTA
reinforce the addiction process in a vicious cycle; opioid receptors contribute dopamine neurons oppose MOR-​stimulating activity at the level of terminals
to all facets of the disease. Animal data (see Supplementary Table 1 for in the NAc and prefrontal cortex18,89 and disrupt behavioural inhibition94.
behavioural models) indicate that MORs drive rewarding properties of opioid KORs in NAc medium spiny neurons differentially gate D1 and D2 neuron
drugs (via direct, on-​target effects) and other drugs of abuse (via indirect, activities and their AMY inputs to negatively regulate motivational
opioid peptide-​mediated effects) in both recreational consumption and processes91. KORs in DRN serotonin (5-HT) neurons promote stress-​induced
binge or intoxication, and that repeated MOR activation leads to reduced reinstatement to cocaine seeking96,97; KORs in excitatory basolateral
drug reward (tolerance) and compensatory adaptations (dependence or amygdala (BL A) neurons projecting to the bed nucleus of stria terminalis
withdrawal symptoms). KOR–Dyn activity is important in the negative affect (BNST) promote anxiety98; and KORs in GABAergic interneurons of the
that characterizes withdrawal as well as in short-​term or prolonged central AMY contribute to excessive alcohol drinking, probably through
abstinence, whereas DORs should limit development of this aversive state. interactions with corticotropin releasing factor systems39,78,83. Finally , MORs
All three opioid receptors probably influence the preoccupation– and KORs are also expressed in brain regions involved in contextual memory
anticipation stage, and are implicated in drug-​biased motivation, habit and executive functions (including the hippocampus (HIP) and frontal cortex
formation and loss of control, but these roles are less well understood. In the (FC))78,103, but the potential implication of these receptors in preoccupation
latter stage, or relapse to drug taking, evidence supports a role for DORs in or anticipation has not been studied so far.

504 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

deletion from GABAergic forebrain neurons (mostly and prolonged abstinence from opiates are both associ­
from D1 neurons), using a Dlx5/6-Cre driver line in ated with negative emotional states that can be mode­
floxed Oprm1 mice, reduced voluntary alcohol drinking lled in rodents44. In this context, MORs in the dorsal
and alcohol CPP64, indicating that this receptor popula- raphe nucleus (DRN), the main serotonergic nucleus,
tion indeed contributes to alcohol reward. Intriguingly, were shown to be crucial for the development of
the latter genetic manipulation did not impair heroin despair-​like behaviour and deficits in social interac-
CPP — presumably because VTA MORs were intact tion in heroin-​abstinent mice74. This finding indicates
in these mice — but did increase seeking behaviour for that DRN MORs regulate serotonergic transmission
heroin and palatable food in SA experiments59, dissoci- and that adaptations of MOR signalling in the DRN in
ating MOR-​mediated hedonic effects from motivational response to chronic opiate exposure may profoundly
regulation. Thus, in addition to driving pleasurable drug alter mood during abstinence. This mechanism is parti­
effects via VTA GABAergic interneurons and, to some cularly interesting in the area of addiction–depression
degree, striatal MSNs (depending on the drug and par- comorbidities75.
adigm), MORs also regulate motivational aspects of
behaviour, specifically at the level of striatal projecting KORs in dysphoria, stress and depression. In contrast
neurons. This notion is consistent with sophisticated to MOR agonists, KOR agonists are strongly aversive.
pharmacological studies that show that MORs not only In humans, these drugs induce an acute dysphoric state,
assign hedonic values to rewards, but also contribute to deteriorate mood and have psychotomimetic effects17,76
decision making through coordinated activity in core (Box 2). These KOR activities may have strong implica-
and shell compartments of the NAc, which integrate tions for several psychiatric disorders, including drug
reward-​related information and guide goal-​directed abuse, affective disorders and, potentially, psychosis18.
actions65. Animal model research has established that the KOR
MORs may also contribute to another facet of and dynorphin, the preferred endogenous KOR ligand,
addiction — that is, impaired self-​control — although form an ‘anti-​reward’ brain system43,77. Notably, in the
this aspect has been less explored. Impulsivity is a critical absence of environmental challenge, the endogenous
susceptibility factor for addiction66, and inhibitory controls KOR–dynorphin system has only subtly dysphoric activ-
decline with disease development39. Strikingly, Oprm1- ity; however, this system is highly responsive to stress78,
knockout mice showed remarkably lower motor and accumulating evidence has shown that addiction
impulsivity in a signalled nose-​poke task67. The notion dramatically activates brain stress systems, includ-
that MORs may facilitate the ‘loss of control’ that ing endogenous KOR signalling43,79–81. In turn, these
characterizes drug abuse is intriguing. Further studies events facilitate the emergence of a negative affective
are required to identify the key MOR-​expressing neuron state — which characterizes withdrawal episodes of
populations responsible for this effect. the addiction cycle as well as protracted abstinence82 —
An important question is whether, in addition to and promote escalation of drug consumption, stress-​
facilitating drug reward and drug seeking in the tra- induced potentiation of drug reward and stress-​induced
ditionally studied mesocorticolimbic circuitry, MORs reinstatement of drug seeking83,84.
could also regulate aversion processing in other brain KORs are expressed at many sites in the addiction
networks. The site of the densest MOR expression in neurocircuitry (Fig. 2c), and a circuit mechanism for
the brain is, in fact, the medial habenula (MHb)68. The KOR-​mediated dysphoria has long been proposed.
MHb and lateral habenula form the habenular com- Behavioural analyses have demonstrated CPP and
plex, which is active in the anticipation of aversive CPA in response to MOR agonists and KOR agonists,
outcomes and has garnered increasing interest in both respectively, and microdialysis studies have revealed
addiction and depression research69,70. As the habenular opposing activities for the two types of drugs on
complex is considered to be a centre for aversive pro- DA release, with MOR agonists disinhibiting VTA DA
cessing and acts as an anti-​reward system — notably, neurons at the level of the cell bodies and KOR ago-
by inhibiting DA neuron activity — the hypothesis that nists inhibiting DA-​releasing terminals in the NAc85,86.
MORs may regulate aversion processing, and perhaps These seminal findings may explain why MORs and
limit aversive responses at the level of MHb neurons, KORs have such contrasting effects (that is, euphoria
is appealing, and investigations along these lines have and dysphoria, respectively), and have established the
just begun71. notion that endogenous activities of the two receptors
Finally, an important consideration is that MORs fine-​tune DA tone to regulate hedonic homeostasis.
are chronically activated under repeated drug expo- Accordingly, deletion of Oprk1 from DA neurons
sure, and even more so under opiates. With chronic reduced CPA to KOR agonists87, had anxiolytic effects
activation, receptor signalling adapts throughout the and enhanced cocaine-​stimulated locomotor activity88.
brain, triggering long-​term molecular modifications Electrophysiology demonstrated that KORs in DA
within and outside the opioid system72; such neuroa­ neurons inhibit terminals not only in the NAc, but also
daptations to chronic opiates are being intensively in the prefrontal cortex (PFC)89. As in the NAc, KORs
studied73. Behavioural consequences of these adapta- reduced DA release locally in the PFC via a presynaptic
Inhibitory controls tions include the development of tolerance (weakening mechanism and thus contributed considerably to KOR
A central component of
executive functions, geared to
drug effects with repeated exposure), and dependence agonist-​induced CPA90. Therefore, KORs in DA neurons
inhibit or delay dominant (which manifests in the form of withdrawal symptoms produce dysphoria via presynaptic inhibition at both
responses to achieve a goal. when the drug is no longer present). Acute withdrawal NAc and PFC sites.

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 505


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

In the NAc, KORs are expressed by D1 and D2 MSNs antagonist into the CeA reduced the increase in alcohol
and at the terminals of glutamatergic projections from SA elicited by chronic intermittent exposure to alcohol
the basolateral amygdala (BLA), and dynorphin is vapours (a procedure designed to trigger escalation of
produced by D1 MSNs. A recent optogenetic study alcohol consumption)99. Slice electrophysiology showed
examined KOR function throughout the entire NAc that KORs tonically inhibit local GABAergic transmis-
microcircuitry and demonstrated that two distinct pre- sion in the CeA via a presynaptic mechanism and has
synaptic KOR-​mediated mechanisms regulate D1 and thus been proposed to regulate the effects of ethanol
D2 MSN activities: one pathway-​specific (affecting BLA there100,101. KORs also interact in a complex manner
inputs to the NAc) and the other local (affecting MSN with the stress hormone corticotropin releasing factor
collaterals)91. This study reveals a mechanism whereby (CRF)102. This KOR–CRF signalling interaction is being
KORs in the NAc dampen excessive MSN activation that increasingly studied for a role in the pro-​addictive effects
is driven by incentive stimuli or stress, thereby limiting of stress, in anxiety disorders and in related psychiatric
reward-​seeking behaviour. Thus, in addition to the role conditions (reviewed elsewhere39,78,83).
of KORs in influencing euphoria–dysphoria balance,
the notion that KORs negatively regulate motivation is DORs in mood and learning. Similar to MORs and
also gaining support. Another optogenetic manipulation KORs, DORs contribute to the development of drug
revealed functional heterogeneity among Pdyn-​positive abuse, although very differently from the two other
neurons (D1 MSNs) in the NAc92. Photostimulation of receptors. Oprd1-knockout mice show intact mor-
these neurons in the dorsal or ventral NAc shell pro- phine SA, intact THC CPP, decreased nicotine SA and
duced approach or avoidance behaviour, respectively, increased alcohol SA (reviewed in refs11,38 and see
in a real-​time place-​preference assay. These opposing Supplementary Table 1). Therefore, contrary to MORs,
effects were both mediated by KORs and were confirmed DORs are not essential for drug reward. Rather, as pro-
by operant photostimulation. These findings add support posed by pharmacological studies, DORs seem to have
to the emerging notion that reward and aversion mech- a complex regulatory role in motivated behaviours65,103.
anisms are highly intermingled within the NAc (as was Unlike Oprm1-knockout mice and Oprk1-knockout
previously shown for the VTA93) and further support the mice, DOR-​null mutants show a remarkable anxiogenic
view that dysfunctional KOR–dynorphin activity in and depressive-​like phenotype, largely confirmed by
the NAc affects motivated behaviours. Another study pharmacological approaches14,104, suggesting that DOR
in mice used an operant test of cognition (in which lower activity normally helps to alleviate the negative mood
rates of response were differentially reinforced) to show associated with acute withdrawal and prolonged absti-
that both stress and KOR activation impair behavioural nence. In support of this notion, Oprd1-knockout mice
inhibition, an effect prevented by Oprk1 gene deletion in undergoing protracted abstinence to heroin show exac-
DA neurons94. Thus, KOR blockade might reduce stress-​ erbated sucrose anhedonia74. Another role of DORs
induced compulsive behaviours that contribute to drug with potential relevance to addiction is the regulation
seeking and addiction. of learning and memory (reviewed elsewhere103,105).
KOR activity modulates other brain regions and DOR-​null mice show impaired spatial learning and,
transmitter systems (in addition to the DA system) that consistently, reduced drug–context association in CPP
are crucial for negative mood and aversive responses paradigms106. These and other observations suggest that
(reviewed in ref.78). First, KORs in the DRN modulate DOR activity in fact enhances drug–context memories
5-HT transmission, similar to how they modulate DA and facilitates relapse. These findings would suggest that
release from VTA neurons. Local application of a KOR the DOR is pro-​addictive; however, by alleviating the
agonist decreased 5-HT release in the DRN95, suggest- low affect associated with withdrawal, DOR activity may
ing that a low DA tone may not be the sole cause for also prevent relapse. In addition, opposing the pheno-
KOR-​mediated dysphoria and that reductions in 5-HT type of MOR-​null mutants, Oprd1-knockout mice show
may also contribute. Accordingly, a KOR antagonist increased motor impulsivity in a signalled nose-​poke
infused in the DRN abolished the CPA induced by task67, suggesting that DOR activity increases inhibitory
systemic KOR agonist administration, and prevented controls. Thus, altogether, the role of DORs in addiction
stress-​induced reinstatement of cocaine CPP, through a is complex.
mechanism that involved p38 mitogen-​activated protein DORs are expressed in all areas subserving reward
kinase (MAPK) in 5-HT neurons and their projections and motivation, mood control, and learning and
Operant photostimulation
to the NAc96,97. Second, amygdala-​based mechanisms memory53,54. The identification of circuit mechanisms
Instrumental conditioning in contribute to KOR-​mediated anxiety. Oprk1 deletion underlying addiction-​r elated functions of DORs
which animals learn to self-​ in the BLA was anxiolytic in mice, and the anxiolytic through cell-​specific genetic approaches is just starting.
administer optogenetic effects produced by photostimulation of the BLA–bed Only one conditional Oprd1-mutant mouse has been
stimulation; used to determine
nucleus of stria terminalis (BNST) pathway were blocked produced, demonstrating that DORs in GABAergic
whether a neuronal population
mediates reward. by systemic administration of a KOR agonist98. The same forebrain neurons mediate the locomotor-​stimulant
study also showed that KORs inhibit excitatory BLA effect of SNC80 (a prototypic DOR agonist), counteract
Escalation terminals in the BNST, and that dynorphin in the BNST D1R agonist-​induced hyperactivity and exert an anxio-
In animal research, extended
thus regulates the activity of BLA–BNST projections genic effect45. Further cell-​targeted genetic studies in the
access to the drug leads to a
daily increase (or escalation) of
to control the amygdala-​centred anxiety circuit. KOR future may address, for example, the intriguing proposed
drug intake, which is suggested activity in the central amygdala (CeA) was also studied role of DORs in stimulus-​based decision-​making at the
to reflect loss of control. in the context of alcohol dependence. Infusion of a KOR level of the BLA–NAc shell circuit65.

506 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Table 1 | Genetic mouse mutants to study opioid receptors in addiction


Mouse line Tool description Findings Refs
Targeting the receptor gene
Knock-​in mice with floxed opioid-​ Dlx5/6-Cre-​mediated conditional knockout • ↑ Motivation for heroin and food SA 59,64

receptor-encoding alleles crossed with of Oprm1 in forebrain GABAergic neurons • ↓ Voluntary alcohol drinking
Cre lines to delete receptor genes in
specific cell populations Dlx5/6-Cre-​mediated conditional knockout • ↓ Anxiety 45

of Oprd1 in forebrain GABAergic neurons • Abolished agonist-​induced locomotor


stimulation
Dat-​Cre-mediated conditional knockout of • Abolished CPA to KOR agonists (U69,593 87,88,186

Oprk1 in DA neurons and U50,488)


• ↑ Locomotor sensitization to cocaine
Knock-​in reporter mice expressing a Oprm1 with carboxy-​terminal mCherry fusion MOR anatomy visualized at brain, neuron 53

functional tagged version of the opioid and subcellular levels


receptor to allow receptor detection
Oprd1 with carboxy-​terminal eGFP fusion • DOR anatomy visualized at brain, neuron 187,188

and subcellular levels


• DOR trafficking in primary neurons
detected in real time
•Receptor trafficking was linked to tolerance
Oprd1 with amino-​terminal HA tag fusion Mouse line characterized 189

combined with a floxed exon 1 for the


conditional deletion of tagged DORs in
targeted neuron populations
Overexpressing the receptor gene
Transgenic lines expressing wild-​type Pdyn-​MOR transgene on an Oprm1-knockout MOR rescue restored morphine reward and 58

opioid receptors background to rescue MOR expression in partly restored remifentanil SA


striatopallidal neurons
Dat-​Cre-dependent viral expression of KORs KOR rescue restored KOR agonist 186

on an Oprk1-knockout background, to rescue (U50,488)-induced CPA


KOR expression in DA neurons
Transgenic lines expressing mutant opioid Light-​activatable opto-​MOR virally expressed Real-​time place preference or avoidance was 190

receptors in GABAergic RMTg and VP neurons dependent on the photostimulated area


Viral KOR-​DREADD expressed in VTA and SN • KOR-​DREADD activation in the midbrain 191,192

or subiculum projections to the NAc (VTA and SN) reduced cocaine-​induced


hyperlocomotion
• KOR-​DREADD activation in subiculum
projections to NAc reduced context-​
induced relapse to alcohol seeking
Targeting opioid-​receptor-expressing or peptide-​expressing neurons
Pomc-​IRES-Crea Cre recombinase expressed in Pomc-​positive Not used in addiction-​related studies 193

neurons that produce β-​endorphin


Penk-​IRES-Crea Cre recombinase expressed in Penk-​positive Not used in addiction-​related studies 193

neurons that produce preproenkephalin


Pdyn-​Cre Cre recombinase expressed in Pdyn-​positive Optogenetic activation of Pdyn-​expressing 92

neurons that produce preprodynorphin cells in the NAc shell produced aversion
(ventral) or preference (dorsal) depending on
the targeted subregion
Oprk1-Cre Cre recombinase expressed in KOR-​positive Mouse line characterized 194

neurons
CPA , conditioned place aversion; DA , dopamine; Dat, gene encoding DA transporter ; Dlx5/6, genes encoding homeobox proteins DLX5 or DLX6; DOR , delta-​
opioid receptor ; DREADD, designer receptor exclusively activated by designer drugs; eGFP, enhanced green fluorescent protein; HA , human influenza
haemagglutinin; IRES, internal ribosome entry site, KOR , kappa-​opioid receptor ; MOR , mu-​opioid receptor ; NAc, nucleus accumbens, Oprd1, gene encoding DOR;
Oprk1, gene encoding KOR; Oprm1, gene encoding MOR; opto-​MOR , optically sensitive MOR; Pdyn, gene encoding preprodynorphin; Penk, gene encoding
preproenkephalin; Pomc, gene encoding proopiomelanocortin; RMTg, rostromedial tegmental nucleus; SA , self-​administration; SN, substantia nigra; VP, ventral
pallidum; VTA , ventral tegmental area. aMice generated by the Allen Institute.

The gap between basic knowledge and treatment tools to manipulate opioid receptors or the specific neu-
strategies. As described above, anatomical and phar- rons that express them are developing rapidly107 (Table 1).
macological studies have shown that MORs, KORs and More circuit mechanisms subserving MOR-​controlled
DORs operate at many sites of addiction circuits, and KOR-​controlled behaviours related to drug abuse will
and genetic approaches have identified several molecu- undoubtedly be discovered, and the contribution of DORs
lar and neuronal mechanisms underlying MOR and KOR to this circuitry — particularly in emotion, motivation
activities at distinct stages of the disease (Fig. 2b,c). Genetic and learning networks — will also be clarified38,103,105.

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 507


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

In principle, opioid receptors should be ideal targets The most common and best-​studied variant associ-
to treat drug abuse. MOR blockade could reduce drug ated with drug addiction is the A118G single-​nucleotide
reward, the motivation to seek drugs and compulsive polymorphism (SNP) in the OPRM1 gene (rs1799971,
behaviour. KOR blockade should limit dysphoric states exon 1), which is frequent in non-​African popula-
associated with stress and drug withdrawal, and help tions (16% in Europeans and 38% in Asians) and less
to prevent stress-​induced relapse. Blocking KORs may frequent among African Americans (3%)120. This non-​
also reduce compulsivity (although it could promote synonymous substitution creates a new CpG site for
drug reward). Finally, DOR activation should reduce DNA methylation and leads to an Asn40Asp substitution
anxiety and limit compulsivity but may also facilitate in the extracellular amino-​terminal domain of the recep-
drug–context learning. Although circuit analyses will nottor protein — two possible reasons for the reduced MOR
expression and signalling detected in cell cultures121 and
directly facilitate drug discovery to treat addiction, they
should provide a better understanding of drug effects andpost-​mortem human tissues122 expressing this variant.
help target development of therapeutic drugs to specific To date, many candidate gene studies have correlated
aspects of this complex disorder, including withdrawal, the presence of this particular OPRM1 SNP with opioid
distinct recovery stages and complete abstinence. dependence, responses to opioid pharmacotherapy,
Today, amazingly few opioid drugs are available and alcoholism and nicotine dependence120 (Table 2).
used in clinical practice. Naltrexone (a MOR antagonist) This particular coding OPRM1 SNP, however, may
is a labelled medication that is used to treat alcohol not be the sole factor involved in these associations,
abuse108 and shows some success for other non-​opioid as other intronic and synonymous coding variants
substance or behavioural addictions109 by reducing may affect gene function at the level of transcription,
MOR-​mediated reward. Methadone (a partial MOR mRNA stability and splicing. Recently, a cis-​expression
agonist) and buprenorphine (a partial MOR agonist quantitative trait loci mapping study identified several
and KOR antagonist) show good efficacy to treat opioid other OPRM1 polymorphisms altering OPRM1 expres-
addiction, essentially as replacement therapies110–112. The
sion in PFC samples from the BrainCloud cohort123 and
KOR antagonist activity of buprenorphine is believed tested their association with risk of heroin addiction124.
to contribute to its therapeutic success, and pure KOR Four SNPs were associated with the condition, and
antagonists are being developed to treat addiction84, this association was replicated only when the A118G
anxiety and depression113. One KOR blocker (JDTic) SNP was in the presence of another SNP in intron 1
failed a phase I trial owing to some adverse effects includ-
(rs3778150), indicating that nearby intronic SNPs may
ing tachycardia, and another KOR blocker (CERC-501) instead underlie the inconsistent associations of the
is currently in trials for smoking cessation, treatment-​main A118G SNP with heroin addiction124 and alcohol
resistant depression and anxiety disorder (phase II addiction125. In another study, the rs3778150-rs1799971
for all) 114. The convergence of animal and human haplotype was also associated with several alcohol-​use
research should lead to an expansion of the therapeutic phenotypes126. More recently, a genome-​wide associ-
potential of opioids for drug abuse disorders. ation study (GWAS) revealed a significant association
between a single SNP (rs73568641) and methadone
Bridging the translational gap maintenance therapy in African Americans but not in
Several lines of research will help to bridge the trans- European Americans; interestingly, the closest gene to
lational gap between the genetic and pharmacological this SNP is OPRM1 (ref.127). Future GWAS using novel
studies in animal models described above and the unbiased machine learning approaches should efficiently
development of novel opioid receptor-​targeting strat- address the complexity of allele combination effects.
egies (that is, innovative drugs and/or personalized The A118G SNP was transposed into mice using two
treatments) to treat addiction in the clinic. Notably, knock-​in approaches, together representing one of the
human research includes genetic association studies and very few attempts to model a human vulnerability gene
neuroimaging research, which unequivocally indicate for a psychiatric disorder. In one knock-​in mouse line,
that opioid receptors — particularly MORs — are the major Oprm1 allele was replaced by a mouse allele
associated with the development of addiction. carrying the SNP corresponding to the A118G SNP in
humans (A112G)128. The other experimental design cre-
Gene variability. Drug addiction is partly heritable; ated two humanized mouse models harbouring either the
Quantitative trait loci therefore, genetic factors contribute to addiction vul- major (A118) or minor (G118) human OPRM1 exon 1
Genomic regions that carry one
or more DNA mutations that
nerability115. Over two decades, association studies (ref.129). Mutant mouse phenotypes were analysed in
correlate with phenotypic have established that all opioid-​encoding or opioid-​ depth, but unfortunately the two lines have not been
variations (for example, receptor-encoding genes are loci associated with the compared in similar paradigms and, as such, phenotype
behaviour, gene expression potential risk of opioid addiction116 or other substance comparison is not straightforward (Table 2). Of note are
and protein levels).
use disorders117. In the case of opioid dependence, for the parallel findings of increased heroin and nicotine SA
Machine learning example, risk associations have been found for var- as well as increased heroin-​induced and alcohol-​induced
A research field in which iants of OPRM1, OPRK1, OPRD1 and PDYN116,118 as DA release in A112G knock-​in mice and humanized
computers learn to extract well as POMC and PENK variants (although the latter A118G mice, respectively. Importantly, two studies
patterns from complex data two associations require replication)116. Genetic and have directly compared human and mouse phenotypes
sets without being explicitly
programmed for this goal.
epigenetic data from studies on the opioid system have using the humanized mouse model129,130, demonstrat-
Helps biologists to build been reviewed very recently in the context of addiction ing the potential for translation from mouse to human
predictions. biomarker discovery119. research. Striatal DA release in response to an alcohol

508 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Table 2 | Effects of the OPRM1 A118G polymorphism in humans and mouse models
Population or model Substance or stimulus Findings Refs
Human studies
Polymorphism ID Opioid 118G allele increased risk of opioid addiction 195–199

rs1799971, exon 1, A118G


No association with or protection from opioid 200

addiction was found


Alcohol 118G allele increased risk of alcohol addiction 195,196,201–204

Only 118G carriers showed a striatal DA 129

response to alcohol
Amphetamine 118A allele was associated with increased 205

euphoria and stimulant effect of amphetamine


Nicotine 118G allele was associated with increased 130,206

pleasurable nicotine effects


Mouse studies
A112G knock-​in mice Opioid 112GG increased heroin SA and striatal DA 207

release
• 112GG reduced morphine-​induced 128

locomotor stimulation
• Female 112GG carriers did not show place
conditioning to morphine
112GG reduced buprenorphine-​induced 208

locomotor stimulation
Social defeat 112GG mice were resilient to social defeat 209

Mouse exon 1 replaced by Opioid 118GG decreased morphine and hydrocodone 210

human A118G exon 1 reward (via intracranial self-​stimulation) and


DA release in the nucleus accumbens
118GG decreased morphine-​induced 210

locomotor stimulation
Place conditioning to morphine was 211

unchanged
Alcohol 118GG increased alcohol-​induced DA releasea 129

118GG increased efficacy of naltrexone and 212

nalmefene in reducing operant alcohol SA


Nicotine 118GG increased nicotine SA in male but not 130

female micea
DA , dopamine; SA , self-​administration. aThese studies directly compared the human and mouse phenotypes.

challenge was enhanced for G-​allele carriers in mice and PET imaging seeks to map and quantify receptor
in humans129, and the pleasurable effects of nicotine in occupancy throughout the brain using radiolabelled
humans or nicotine SA in mice were increased in males, tracers. Receptor bioavailability, also known as binding
but not females, of each species130. potential (BP), varies depending on receptor expres-
The mouse data are certainly complex and do not sion and localization, levels of endogenous ligand and
exactly match the human findings. Nevertheless, the the binding of exogenous drugs. For opioid receptors,
hypothesized reductions in MOR availability and most reports describe MOR BP using 11C-​carfentanil,
function, in cells and throughout the brain of A118G a highly selective, high-​affinity MOR agonist, whereas
carriers, seem to modify the neurobiology of drug DORs or KORs remain difficult to study owing to the
reward. Altered reward processing may be one of the paucity of suitable tracers132. Overall, MOR bioavailabil-
mechanisms underlying the reported higher risk of ity is modified by exposure to drugs of abuse (including
addiction in these individuals. opiates, cocaine, alcohol and nicotine), drug craving
and dependence, opioid pharmacotherapy, affective
Neuroimaging. Non-​invasive neuroimaging techniques, experiences and pain states (reviewed in refs119,132).
Receptor bioavailability
Also known as receptor including positron emission tomography (PET) and Recently, 11C-​carfentanil PET imaging revealed that
binding. The quantity of MRI, have been used for decades to explore neurotrans- MOR BP is reduced in the frontal cortex of people who
radiotracer that binds to its mitter system activities and whole-​brain functioning in smoke cigarettes and correlates with cigarette liking
target receptor in positron humans and are used to evaluate, for example, the efficacy and wanting, suggesting a MOR-​mediated activation
emission tomography imaging.
Depends on receptor levels
of addiction treatments131. Rodent neuroimaging using mechanism for nicotine reward and dependence133. In a
and occupancy by endogenous similar approaches has lagged behind owing to limited separate study, a short overnight abstinence period was
ligands. resolution but is gaining interest as methods improve. sufficient to decrease MOR BP in smokers compared

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 509


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

with non-​smokers, and the reduction of MOR availabil- to characterize long-​t erm modifications of brain
ity in the basal ganglia was correlated with craving and states with chronic drug exposure. A set of 21 studies
the severity of nicotine dependence134. Moreover, the assessing individuals with a history of heroin use was
A118G SNP was associated with lower baseline MOR recently meta-​analysed. All of these studies pointed
BP in the NAc and amygdala of smokers134. Another to alterations in connectivity that converge to pro-
autoradiographic study showed a reduction of MOR mote dysfunctional decision-​making and indicated
binding sites in post-​mortem striatal tissue from individ- that network modifications increase with the surveyed
uals with alcoholism, paralleling low MOR BP in patients duration of heroin exposure and last for a long time after
with alcoholism. PET imaging also revealed that a reduc- becoming abstinent146. Heroin-​abstinent individuals
tion in MOR availability in detoxified alcohol-​dependent also all showed differences in the strength of functional
patients may be associated with a higher risk of relapse135. connectivity in brain systems involved in reward pro-
Intriguingly, recent PET studies have also revealed the cessing, stress responses and impulsivity147. A recent
role of MORs in natural rewards and behavioural addic- study analysed changes in connectivity in individuals
tions. Lower MOR BP has been consistently reported who were formerly addicted to heroin and were under
among individuals showing pathological gambling or methadone maintenance therapy. Effective connectivity
binge eating136,137, and even in obese individuals who was measured for four networks (reward, motivation,
self-​report food addiction138. High-​intensity training, but memory and cognitive control). Whereas reward and
not moderate exercise, decreased MOR BP in fronto­ cognitive control centres were more weakly connected
limbic regions related to reward and emotional process- with the rest of the brain, learning-​related and memory-​
ing and correlated with euphoria, consistent in this case related centres were more strongly connected than in
with the notion that endogenous opioid peptide release heroin-​naive individuals148. These findings are particu-
may reduce MOR availability139. Along these lines, social larly interesting with regards to bridging human and
interactions such as social touch140 and social laughter141 animal research as similar processes are modelled in
modulate MOR availability. Although the interpretation preclinical research (Fig. 2b).
of these two studies seems complex, the link between Finally, fMRI is now being developed in mice to
social behaviour and opioidergic activity has been well study MOR activation and function, with the unique
established in humans. More generally, PET-​based evi- advantage of including Oprm1-knockout groups in the
dence suggests that MOR-​mediated endogenous opioid experimental design to isolate on-​target MOR-​mediated
activity facilitates approach-​oriented emotions (such as effects. phMRI analysis of oxycodone effects in awake
anger and pleasure), inhibits avoidance-​oriented emo- mice identified an activation pattern149 with anatomi-
tions (such as fear or sadness) and modulates affiliative cal similarities to the effects of morphine in humans145.
behaviours in humans142. Human studies therefore con- Furthermore, an unbiased rs-​fMRI analysis comparing
verge well with animal research to demonstrate that the whole-​brain activity in anaesthetized mice revealed that
MOR is important in reward–aversion processing and Oprm1-knockout mice show perturbed resting-​state
promotes sociability and resilience to stress and that functional connectivity, with predominant alteration of
changes in MOR-​mediated opioidergic activity may the connectivity of reward and aversion centres34. These
therefore contribute considerably to the development recent studies indicate that fMRI is now feasible in
and maintenance of addiction. mouse genetic mutants with a resolution comparable to
Functional MRI (fMRI) is being increasingly used that in human imaging and open exciting new perspec-
to assess brain activation patterns and functional tives for connectome genetics29, therapeutic design143
connectivity, and a variant of fMRI known as phar- and the search for biomarkers119 in opioid research.
macological MRI (phMRI)143 has allowed the mapping
of the acute effects of opioids in the human brain. In Conclusions
healthy, opioid-​naive individuals, a morphine challenge Research in humans and animal models has firmly
increased and decreased blood-​oxygen-level-​dependent established that opioid receptors are central to reward
(BOLD) signals in the NAc (the key reward centre) and aversion processing in the normal brain and con-
and the periaqueductal grey (which processes pain), tribute to the development of addiction. The supporting
respectively144. In a subsequent study, buprenorphine literature is huge, and this Review focuses on recent pro-
dose-​dependently increased BOLD signals in several gress and promises for innovative therapeutic discovery
brain structures (anterior cingulate, frontal cortex, cau- and therapeutic strategies as well as the translational
date putamen, insula and periaqueductal grey), and the potential of opioid research in addiction.
pattern was similar in healthy human participants and in In summary, MORs, DORs and KORs contribute
awake, naive rats, demonstrating the translatability of the to distinct and specific aspects of the disease: MORs
phMRI approach145. In the future, refined drug phMRI promote recreational drug use (including opioids and
‘signatures’ should guide dose selection for therapeutic others) and adapt to chronic activation (leading to tol-
development and clinical trials and provide brain-​level erance and dependence); KORs enable and sustain aver-
mechanistic insights into the incompletely understood sive states of withdrawal and abstinence; DORs improve
and variable in vivo effects of biased MOR agonists that mood states and facilitate context learning; and all three
Effective connectivity are currently being developed (Box 1) or MOR agonists receptors modulate motivation. That both MOR and
In functional MRI, a measure of
the influence of one brain
already used in the clinic. KOR activities drive the onset, progression and main-
region on the activity of Resting-​state fMRI (rs-​fMRI) has also been used tenance of addiction is well recognized, whereas the
another brain region. to investigate brain network activities at rest and contribution of DORs remains less straightforward.

510 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

In this Review, we have tentatively addressed three receptors that may help the interpretation of the effects
questions. First, can we kill pain without addiction of compounds in humans and expand the breadth of
using MOR-​targeting opiates? Structural and signalling indications for the very few therapies that exist as well
receptor biology have brought fascinating answers to this as future opioid-​targeting agents for treating addiction.
long-​standing question and have guided the creation of Third, can we bridge opioid research in the field of drug
entirely novel chemical entities, and system-​level animal abuse in animal models with that in humans? Most
studies and clinical trials will ultimately reveal the poten- of such research (genetic, epigenetic and neuroimag-
tial of these novel compounds. Second, how do opioid ing) in humans has been focused on MORs, although
receptors operate with the neurocircuitry of addiction? research into KORs is increasing. The advent of human-
The identification of neurons and circuits subserving ized mouse genetic models and rodent neuroimaging is
these activities is rapidly progressing as more genetic expected to integrate molecular and circuit mechanisms
tools become available. Whether recent basic research into clinical knowledge and to enhance the potential for
efforts in this field will improve treatment strategies in personalized addiction treatment.
the future is not clear. However, preclinical circuitry
research has revealed less well-​known roles for opioid Published online 22 June 2018

1. Brownstein, M. J. A brief history of opiates, opioid 19. Koob, G. F. The dark side of emotion: the addiction 32. Hu, H. Reward and aversion. Annu. Rev. Neurosci. 39,
peptides, and opioid receptors. Proc. Natl Acad. Sci. perspective. Eur. J. Pharmacol. 753, 73–87 (2015). 297–324 (2016).
USA 90, 5391–5393 (1993). 20. Kobilka, B. The structural basis of G-​protein-coupled 33. Borsook, D. et al. Reward-​aversion circuitry in
2. World Health Organization. Curbing prescription receptor signaling (Nobel Lecture). Angew. Chem. Int. analgesia and pain: implications for psychiatric
opioid dependency. WHO http://www.who.int/bulletin/ Ed Engl. 52, 6380–6388 (2013). disorders. Eur. J. Pain 11, 7–20 (2007).
volumes/95/5/17-020517/en/ (2017). 21. Audet, M. & Bouvier, M. Restructuring G-​protein- 34. Mechling, A. E. et al. Deletion of the mu opioid
3. Kolodny, A. et al. The prescription opioid and heroin coupled receptor activation. Cell 151, 14–23 (2012). receptor gene in mice reshapes the reward–aversion
crisis: a public health approach to an epidemic of 22. Manglik, A. et al. Crystal structure of the micro-​opioid connectome. Proc. Natl Acad. Sci. USA 113,
addiction. Annu. Rev. Publ. Health 36, 559–574 receptor bound to a morphinan antagonist. Nature 11603–11608 (2016).
(2015). 485, 321–326 (2012). This study analyses functional connectivity by
4. Volkow, N. D. & McLellan, A. T. Opioid abuse in This study reports the atomic structure of the MOR non-invasive MRI in live mutant mice and reveals a
chronic pain — misconceptions and mitigation solved by X-​ray crystallography and is published fingerprint of Oprm1 activity over the whole brain,
strategies. N. Engl. J. Med. 374, 1253–1263 back to back with the first atomic structures of the opening the way to connectome genetics in animal
(2016). three other opioid and opioid-​like receptors, all research.
5. Voon, P., Karamouzian, M. & Kerr, T. Chronic pain and bound to antagonists. 35. Navratilova, E., Atcherley, C. W. & Porreca, F. Brain
opioid misuse: a review of reviews. Subst. Abuse Treat. 23. Granier, S. et al. Structure of the delta-​opioid circuits encoding reward from pain relief. Trends
Prev. Policy 12, 36 (2017). receptor bound to naltrindole. Nature 485, Neurosci. 38, 741–750 (2015).
6. Compton, W. M., Jones, C. M. & Baldwin, G. T. 400–404 (2012). 36. Lutz, P. E. & Kieffer, B. L. The multiple facets of opioid
Relationship between nonmedical prescription-​opioid This study reports the atomic structure of the DOR receptor function: implications for addiction. Curr.
use and heroin use. N. Engl. J. Med. 374, 154–163 solved by X-​ray crystallography and is published Opin. Neurobiol. 23, 473–479 (2013).
(2016). back to back with the first atomic structures of the 37. Eisenberger, N. I. The pain of social disconnection:
7. Suzuki, J. & El-​Haddad, S. A review: fentanyl and three other opioid and opioid-​like receptors, all examining the shared neural underpinnings of physical
non-pharmaceutical fentanyls. Drug Alcohol Depend. bound to antagonists. and social pain. Nat. Rev. Neurosci. 13, 421–434
171, 107–116 (2017). 24. Wu, H. et al. Structure of the human kappa-​opioid (2012).
8. Al-​Hasani, R. & Bruchas, M. R. Molecular mechanisms receptor in complex with JDTic. Nature 485, 38. Lutz, P. E. & Kieffer, B. L. Opioid receptors: distinct
of opioid receptor-​dependent signaling and behavior. 327–332 (2012). roles in mood disorders. Trends Neurosci. 36,
Anesthesiology 115, 1363–1381 (2011). This study reports the atomic structure of the KOR 195–206 (2013).
9. Bodnar, R. J. Endogenous opiates and behavior: solved by X-​ray crystallography and is published 39. Koob, G. F. & Volkow, N. D. Neurobiology of addiction:
2015. Peptides 88, 126–188 (2017). back to back with the first atomic structures of the a neurocircuitry analysis. Lancet Psychiatry 3,
10. Kieffer, B. L. & Gaveriaux-​Ruff, C. Exploring the opioid three other opioid and opioid-​like receptors, all 760–773 (2016).
system by gene knockout. Prog. Neurobiol. 66, bound to antagonists. This seminal review summarizes current knowledge
285–306 (2002). 25. Thompson, A. A. et al. Structure of the nociceptin/ on animal addiction research, including conceptual
11. Charbogne, P., Kieffer, B. L. & Befort, K. 15 years of orphanin FQ receptor in complex with a peptide frameworks, behavioural models, brain circuitry
genetic approaches in vivo for addiction research: mimetic. Nature 485, 395–399 (2012). and neurobiology.
opioid receptor and peptide gene knockout in mouse This study reports the atomic structure of the NOR 40. Koob, G. F. & Volkow, N. D. Neurocircuitry of
models of drug abuse. Neuropharmacology 76, solved by X-​ray crystallography and is published addiction. Neuropsychopharmacology 35, 217–238
204–217 (2014). back to back with the first atomic structures of the (2010).
12. Matthes, H. W. et al. Loss of morphine-​induced three other opioid and opioid-​like receptors, all 41. Baler, R. D. & Volkow, N. D. Drug addiction: the
analgesia, reward effect and withdrawal symptoms in bound to antagonists. neurobiology of disrupted self-​control. Trends Mol.
mice lacking the mu-​opioid-receptor gene. Nature 26. Manglik, A. et al. Structure-​based discovery of opioid Med. 12, 559–566 (2006).
383, 819–823 (1996). analgesics with reduced side effects. Nature 537, 42. Belin, D., Belin-​Rauscent, A., Murray, J. E. &
This is a genetic demonstration that both 185–190 (2016). Everitt, B. J. Addiction: failure of control over
therapeutic and unwanted morphine effects are This study reports computational screening based maladaptive incentive habits. Curr. Opin. Neurobiol.
mediated by a single receptor protein (MOR). on the MOR structure, allowing the discovery of 23, 564–572 (2013).
13. Filliol, D. et al. Mice deficient for delta- and novel agonist chemotypes unrelated to known 43. Koob, G. F. et al. Addiction as a stress surfeit disorder.
mu-​opioid receptors exhibit opposing alterations of opioids and leading to a Gi-​biased drug with Neuropharmacology 76, 370–382 (2014).
emotional responses. Nat. Genet. 25, 195–200 unusual and promising biological properties 44. Goeldner, C. et al. Impaired emotional-​like behavior
(2000). towards safer analgesics. and serotonergic function during protracted
14. Pradhan, A. A., Befort, K., Nozaki, C., Gaveriaux-​Ruff, C. 27. Bradley, S. J. & Tobin, A. B. Design of next-​generation abstinence from chronic morphine. Biol. Psychiatry
& Kieffer, B. L. The delta opioid receptor: an evolving G protein-​coupled receptor drugs: linking novel 69, 236–244 (2011).
target for the treatment of brain disorders. Trends pharmacology and in vivo animal models. Annu. Rev. 45. Chu Sin Chung, P. et al. A novel anxiogenic role for the
Pharmacol. Sci. 32, 581–590 (2011). Pharmacol. Toxicol. 56, 535–559 (2016). delta opioid receptor expressed in GABAergic
15. Chu Sin Chung, P. & Kieffer, B. L. Delta opioid 28. Kim, C. K., Adhikari, A. & Deisseroth, K. Integration of forebrain neurons. Biol. Psychiatry 77, 404–415
receptors in brain function and diseases. Pharmacol. optogenetics with complementary methodologies in (2015).
Ther. 140, 112–120 (2013). systems neuroscience. Nat. Rev. Neurosci. 18, 46. Contet, C., Kieffer, B. L. & Befort, K. Mu opioid
16. Spahn, V. & Stein, C. Targeting delta opioid receptors 222–235 (2017). receptor: a gateway to drug addiction. Curr. Opin.
for pain treatment: drugs in phase I and II clinical 29. Van Essen, D. C. Cartography and connectomes. Neurobiol. 14, 370–378 (2004).
development. Expert Opin. Investig. Drugs 26, Neuron 80, 775–790 (2013). 47. Badiani, A., Belin, D., Epstein, D., Calu, D. &
155–160 (2017). 30. Bruehl, S. et al. Personalized medicine and Shaham, Y. Opiate versus psychostimulant addiction:
17. Pfeiffer, A., Brantl, V., Herz, A. & Emrich, H. M. opioid analgesic prescribing for chronic pain: the differences do matter. Nat. Rev. Neurosci. 12,
Psychotomimesis mediated by kappa opiate receptors. opportunities and challenges. J. Pain 14, 685–700 (2011).
Science 233, 774–776 (1986). 103–113 (2013). 48. Becker, J. A. J., Kieffer, B. L. & Le Merrer, J.
18. Tejeda, H. A., Shippenberg, T. S. & Henriksson, R. 31. Luo, S. X. & Levin, F. R. Towards precision addiction Differential behavioral and molecular alterations
The dynorphin/kappa-​opioid receptor system and its treatment: new findings in co-​morbid substance use upon protracted abstinence from cocaine versus
role in psychiatric disorders. Cell. Mol. Life Sci. 69, and attention-​deficit hyperactivity disorders. Curr. morphine, nicotine, THC and alcohol. Addict. Biol. 22,
857–896 (2012). Psychiatry Rep. 19, 14 (2017). 1205–1217 (2017).

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 511


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

49. Moles, A., Kieffer, B. L. & D’Amato, F. R. Deficit in of opioid tolerance beyond desensitization and 96. Land, B. B. et al. Activation of the kappa opioid receptor
attachment behavior in mice lacking the mu-​opioid downregulation. Trends Pharmacol. Sci. 37, 963–976 in the dorsal raphe nucleus mediates the aversive
receptor gene. Science 304, 1983–1986 (2004). (2016). effects of stress and reinstates drug seeking. Proc. Natl
This study demonstrates, using a genetic approach, 74. Lutz, P. E. et al. Distinct mu, delta, and kappa opioid Acad. Sci. USA 106, 19168–19173 (2009).
that the MOR is essential for social bonding very receptor mechanisms underlie low sociability and 97. Bruchas, M. R. et al. Selective p38α MAPK deletion in
early on (in 4–8 day old pups), providing key depressive-​like behaviors during heroin abstinence. serotonergic neurons produces stress resilience in
evidence for a MOR contribution to process natural Neuropsychopharmacology 39, 2694–2705 (2014). models of depression and addiction. Neuron 71,
rewards and a strong premise to the rising interest 75. Ng, E., Browne, C. J., Samsom, J. N. & Wong, A. H. C. 498–511 (2011).
in MOR function in the social brain. Depression and substance use comorbidity: what we 98. Crowley, N. A. et al. Dynorphin controls the gain of an
50. Becker, J. A. et al. Autistic-​like syndrome in mu opioid have learned from animal studies. Am. J. Drug Alcohol amygdalar anxiety circuit. Cell Rep. 14, 2774–2783
receptor null mice is relieved by facilitated mGluR4 Abuse 43, 456–474 (2017). (2016).
activity. Neuropsychopharmacology 39, 2049–2060 76. Ranganathan, M. et al. Dose-​related behavioral, 99. Kissler, J. L. & Walker, B. M. Dissociating motivational
(2014). subjective, endocrine, and psychophysiological effects from physiological withdrawal in alcohol dependence:
51. Papaleo, F., Kieffer, B. L., Tabarin, A. & Contarino, A. of the kappa opioid agonist Salvinorin A in humans. role of central amygdala kappa-​opioid receptors.
Decreased motivation to eat in mu-​opioid Biol. Psychiatry 72, 871–879 (2012). Neuropsychopharmacology 41, 560–567 (2016).
receptor-deficient mice. Eur. J. Neurosci. 25, 77. Koob, G. F. & Le Moal, M. Addiction and the brain 100. Kang-​Park, M., Kieffer, B. L., Roberts, A. J., Siggins, G. R.
3398–3405 (2007). antireward system. Annu. Rev. Psychol. 59, 29–53 & Moore, S. D. Kappa-​opioid receptors in the central
52. Skoubis, P. D., Matthes, H. W., Walwyn, W. M., (2008). amygdala regulate ethanol actions at presynaptic
Kieffer, B. L. & Maidment, N. T. Naloxone fails to 78. Crowley, N. A. & Kash, T. L. Kappa opioid receptor GABAergic sites. J. Pharmacol. Exp. Ther. 346,
produce conditioned place aversion in mu-​opioid signaling in the brain: circuitry and implications for 130–137 (2013).
receptor knock-​out mice. Neuroscience 106, treatment. Prog. Neuropsychopharmacol. Biol. 101. Gilpin, N. W., Roberto, M., Koob, G. F. & Schweitzer, P.
757–763 (2001). Psychiatry 62, 51–60 (2015). Kappa opioid receptor activation decreases inhibitory
53. Erbs, E. et al. A mu-​delta opioid receptor brain atlas 79. Land, B. B. et al. The dysphoric component of stress is transmission and antagonizes alcohol effects in rat
reveals neuronal co-​occurrence in subcortical encoded by activation of the dynorphin kappa-​opioid central amygdala. Neuropharmacology 77, 294–302
networks. Brain Struct. Funct. 220, 677–702 (2015). system. J. Neurosci. 28, 407–414 (2008). (2014).
54. Le Merrer, J., Becker, J. A., Befort, K. & Kieffer, B. L. 80. Zhou, Y. & Kreek, M. J. Alcohol: a stimulant activating 102. Kang-​Park, M., Kieffer, B. L., Roberts, A. J., Siggins, G. R.
Reward processing by the opioid system in the brain. brain stress responsive systems with persistent neuro­ & Moore, S. D. Interaction of CRF and kappa opioid
Physiol. Rev. 89, 1379–1412 (2009). adaptation. Neuropharmacology 87, 51–58 (2014). systems on GABAergic neurotransmission in the
55. Johnson, S. W. & North, R. A. Opioids excite 81. Massaly, N., Moron, J. A. & Al-​Hasani, R. A. Trigger mouse central amygdala. J. Pharmacol. Exp. Ther.
dopamine neurons by hyperpolarization of local for opioid misuse: chronic pain and stress dysregulate 355, 206–211 (2015).
interneurons. J. Neurosci. 12, 483–488 (1992). the mesolimbic pathway and kappa opioid system. 103. Klenowski, P., Morgan, M. & Bartlett, S. E. The role of
56. Wise, R. A. & Rompre, P. P. Brain dopamine and Front. Neurosci. 10, 480 (2016). delta-​opioid receptors in learning and memory
reward. Annu. Rev. Psychol. 40, 191–225 (1989). 82. Lalanne, L. et al. Kappa opioid receptor antagonism underlying the development of addiction. Br. J.
57. Fields, H. L. & Margolis, E. B. Understanding opioid and chronic antidepressant treatment have beneficial Pharmacol. 172, 297–310 (2015).
reward. Trends Neurosci. 38, 217–225 (2015). activities on social interactions and grooming deficits 104. Gendron, L., Cahill, C. M., von Zastrow, M., Schiller, P. W.
58. Cui, Y. et al. Targeted expression of mu-​opioid receptors during heroin abstinence. Addict. Biol. 22, & Pineyro, G. Molecular pharmacology of delta-​opioid
in a subset of striatal direct-​pathway neurons restores 1010–1021 (2017). receptors. Pharmacol. Rev. 68, 631–700 (2016).
opiate reward. Nat. Neurosci. 17, 254–261 (2014). 83. Bruchas, M. R., Land, B. B. & Chavkin, C. The 105. Pellissier, L. P., Pujol, C. N., Becker, J. A. &
59. Charbogne, P. et al. Mu opioid receptors in gamma-​ dynorphin/kappa opioid system as a modulator of Le Merrer, J. Delta opioid receptors: learning and
aminobutyric acidergic forebrain neurons moderate stress-​induced and pro-​addictive behaviors. Brain Res. motivation. Handb Exp. Pharmacol. https://doi.org/
motivation for heroin and palatable food. Biol. 1314, 44–55 (2010). 10.1007/164_2016_89 (2016).
Psychiatry 81, 778–788 (2017). 84. Chavkin, C. & Koob, G. F. Dynorphin, dysphoria, and 106. Le Merrer, J. et al. Deletion of the delta opioid
60. Ettenberg, A., Pettit, H. O., Bloom, F. E. & Koob, G. F. dependence: the stress of addiction. receptor gene impairs place conditioning but
Heroin and cocaine intravenous self-​administration in Neuropsychopharmacology 41, 373–374 (2016). preserves morphine reinforcement. Biol. Psychiatry
rats: mediation by separate neural systems. 85. Spanagel, R., Herz, A. & Shippenberg, T. S. Opposing 69, 700–703 (2011).
Psychopharmacology 78, 204–209 (1982). tonically active endogenous opioid systems modulate 107. Bruchas, M. R. & Roth, B. L. New technologies for
61. Hnasko, T. S., Sotak, B. N. & Palmiter, R. D. Morphine the mesolimbic dopaminergic pathway. Proc. Natl elucidating opioid receptor function. Trends
reward in dopamine-​deficient mice. Nature 438, Acad. Sci. USA 89, 2046–2050 (1992). Pharmacol. Sci. 37, 279–289 (2016).
854–857 (2005). 86. Bals-​Kubik, R., Ableitner, A., Herz, A. & 108. Koob, G. F. & Mason, B. J. Existing and future drugs
62. Laviolette, S. R. & van der Kooy, D. GABAA receptors Shippenberg, T. S. Neuroanatomical sites mediating for the treatment of the dark side of addiction. Annu.
signal bidirectional reward transmission from the the motivational effects of opioids as mapped by Rev. Pharmacol. Toxicol. 56, 299–322 (2016).
ventral tegmental area to the tegmental the conditioned place preference paradigm in rats. 109. Aboujaoude, E. & Salame, W. O. Naltrexone: a pan-
pedunculopontine nucleus as a function of opiate J. Pharmacol. Exp. Ther. 264, 489–495 (1993). addiction treatment? CNS Drugs 30, 719–733 (2016).
state. Eur. J. Neurosci. 20, 2179–2187 (2004). 87. Chefer, V. I., Backman, C. M., Gigante, E. D. & 110. Schuckit, M. A. Treatment of opioid-​use disorders.
63. Narayanan, S. et al. Endogenous opioids mediate Shippenberg, T. S. Kappa opioid receptors on N. Engl. J. Med. 375, 357–368 (2016).
basal hedonic tone independent of dopamine dopaminergic neurons are necessary for 111. Volkow, N. D. & Skolnick, P. New medications for
D-1 or D-2 receptor activation. Neuroscience 124, kappa-mediated place aversion. substance use disorders: challenges and opportunities.
241–246 (2004). Neuropsychopharmacology 38, 2623–2631 (2013). Neuropsychopharmacology 37, 290–292 (2012).
64. Ben Hamida, S., Boulos, L. J., McNicholas, M., 88. Van’t Veer, A. et al. Ablation of kappa-​opioid receptors 112. Ayanga, D., Shorter, D. & Kosten, T. R. Update on
Charbogne, P. & Kieffer, B. L. Mu opioid receptors in from brain dopamine neurons has anxiolytic-​like pharmacotherapy for treatment of opioid use disorder.
GABAergic neurons of the forebrain promote alcohol effects and enhances cocaine-​induced plasticity. Expert Opin. Pharmacother. 17, 2307–2318 (2016).
reward and drinking. Addict. Biol. https://doi.org/ Neuropsychopharmacology 38, 1585–1597 (2013). 113. Carlezon, W. A. Jr & Krystal, A. D. Kappa-​opioid
10.1111/adb.12576 (2017). 89. Margolis, E. B. et al. Kappa opioids selectively control antagonists for psychiatric disorders: from bench to
65. Laurent, V., Morse, A. K. & Balleine, B. W. The role dopaminergic neurons projecting to the prefrontal clinical trials. Depress Anxiety 33, 895–906 (2016).
of opioid processes in reward and decision-​making. cortex. Proc. Natl Acad. Sci. USA 103, 2938–2942 114. Helal, M. A., Habib, E. S. & Chittiboyina, A. G.
Br. J. Pharmacol. 172, 449–459 (2015). (2006). Selective kappa opioid antagonists for treatment of
66. Dalley, J. W. & Robbins, T. W. Fractionating 90. Tejeda, H. A. et al. Prefrontal cortical kappa-​opioid addiction, are we there yet? Eur. J. Med. Chem. 141,
impulsivity: neuropsychiatric implications. Nat. Rev. receptor modulation of local neurotransmission and 632–647 (2017).
Neurosci. 18, 158–171 (2017). conditioned place aversion. Neuropsychopharmacology 115. Goldman, D., Oroszi, G. & Ducci, F. The genetics of
67. Olmstead, M. C., Ouagazzal, A. M. & Kieffer, B. L. 38, 1770–1779 (2013). addictions: uncovering the genes. Nat. Rev. Genet. 6,
Mu and delta opioid receptors oppositely regulate 91. Tejeda, H. A. et al. Pathway- and cell-​specific kappa-​ 521–532 (2005).
motor impulsivity in the signaled nose poke task. opioid receptor modulation of excitation-​inhibition 116. Reed, B., Butelman, E. R., Yuferov, V., Randesi, M. &
PLoS ONE 4, e4410 (2009). balance differentially gates D1 and D2 accumbens Kreek, M. J. Genetics of opiate addiction. Curr.
68. Gardon, O. et al. Expression of mu opioid receptor in neuron activity. Neuron 93, 147–163 (2017). Psychiatry Rep. 16, 504 (2014).
dorsal diencephalic conduction system: new insights 92. Al-​Hasani, R. et al. Distinct subpopulations of nucleus 117. Levran, O., Yuferov, V. & Kreek, M. J. The genetics of
for the medial habenula. Neuroscience 277, accumbens dynorphin neurons drive aversion and the opioid system and specific drug addictions. Hum.
595–609 (2014). reward. Neuron 87, 1063–1077 (2015). Genet. 131, 823–842 (2012).
69. Hikosaka, O. The habenula: from stress evasion to This report reveals regional heterogeneity within a 118. Butelman, E. R., Yuferov, V. & Kreek, M. J. Kappa-​
value-​based decision-​making. Nat. Rev. Neurosci. 11, well-​known opioid peptide-​expressing brain area opioid receptor/dynorphin system: genetic and
503–513 (2010). for the first time, as dynorphin-​expressing cells in pharmacotherapeutic implications for addiction.
70. Boulos, L. J., Darcq, E. & Kieffer, B. L. Translating the either the dorsal or ventral part of the NAc shell Trends Neurosci. 35, 587–596 (2012).
habenula-​from rodents to humans. Biol. Psychiatry drive opposing behaviours (that is, reward or 119. Belzeaux, R., Lalanne, L., Kieffer, B. L. & Lutz, P. E.
81, 296–305 (2017). aversion, respectively). Focusing on the opioid system for addiction
71. Boulos, L. J. Mu opioid receptors in the habenula: 93. Lammel, S. et al. Input-​specific control of reward and biomarker discovery. Trends Mol. Med. 24, 206–220
dissecting reward and aversion in addiction. Program aversion in the ventral tegmental area. Nature 491, (2018).
No. 78.11. 2016 Neuroscience Meeting Planner. San 212–217 (2012). 120. Crist, R. C. & Berrettini, W. H. Pharmacogenetics of
Diego, CA: Society for Neuroscience, 2016. 94. Abraham, A. D. et al. Kappa-​opioid receptor activation OPRM1. Pharmacol. Biochem. Behav. 123, 25–33
72. Williams, J. T. et al. Regulation of mu-​opioid receptors: in dopamine neurons disrupts behavioral inhibition. (2014).
desensitization, phosphorylation, internalization, and Neuropsychopharmacology 43, 362–372 (2018). 121. Kroslak, T. et al. The single nucleotide polymorphism
tolerance. Pharmacol. Rev. 65, 223–254 (2013). 95. Tao, R. & Auerbach, S. B. Mu-​opioids disinhibit and A118G alters functional properties of the human
73. Cahill, C. M., Walwyn, W., Taylor, A. M. W., kappa-​opioids inhibit serotonin efflux in the dorsal mu opioid receptor. J. Neurochem. 103, 77–87
Pradhan, A. A. A. & Evans, C. J. Allostatic mechanisms raphe nucleus. Brain Res. 1049, 70–79 (2005). (2007).

512 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

122. Oertel, B. G. et al. A common human μ-​opioid receptor This is a study using phMRI to compare the effect of This article reports a highly innovative approach to
genetic variant diminishes the receptor signaling efficacy buprenorphine on the BOLD signal in both humans design safer opioid pain killers, in which ligand
in brain regions processing the sensory information of and rats and reveals a similar activity pattern across docking to a ‘pathological’ (acidic) receptor
pain. J. Biol. Chem. 284, 6530–6535 (2009). pain-​related regions in the two species. structure identifies a MOR agonist acting at the
123. Colantuoni, C. et al. Temporal dynamics and genetic 146. Fareed, A. et al. Effect of heroin use on changes of receptor only under conditions of inflammation,
control of transcription in the human prefrontal brain functions as measured by functional magnetic leaving other receptors at rest.
cortex. Nature 478, 519–523 (2011). resonance imaging, a systematic review. J. Addict. Dis. 169. Livingston, K. E. & Traynor, J. R. Allostery at opioid
124. Hancock, D. B. et al. Cis-​expression quantitative trait 36, 105–116 (2017). receptors: modulation with small molecule ligands. Br. J.
loci mapping reveals replicable associations with 147. Ieong, H. F. & Yuan, Z. Resting-​state neuroimaging and Pharmacol. https://doi.org/10.1111/bph.13823 (2017).
heroin addiction in OPRM1. Biol. Psychiatry 78, neuropsychological findings in opioid use disorder 170. Burford, N. T. et al. Discovery of positive allosteric
474–484 (2015). during abstinence: a review. Front. Hum. Neurosci. 11, modulators and silent allosteric modulators of the
125. Kong, X. et al. Lack of associations of the opioid 169 (2017). mu-opioid receptor. Proc. Natl Acad. Sci. USA 110,
receptor mu 1 (OPRM1) A118G polymorphism 148. Zhang, Y. et al. Granger causality reveals a dominant 10830–10835 (2013).
(rs1799971) with alcohol dependence: review and role of memory circuit in chronic opioid dependence. 171. Livingston, K. E. & Traynor, J. R. Disruption of the Na+
meta-​analysis of retrospective controlled studies. BMC Addict. Biol. 22, 1068–1080 (2017). ion binding site as a mechanism for positive allosteric
Med. Genet. 18, 120 (2017). 149. Moore, K. et al. BOLD imaging in awake wild-​type and modulation of the mu-​opioid receptor. Proc. Natl
126. Otto, J. M., Gizer, I. R., Deak, J. D., Fleming, K. A. & mu-​opioid receptor knock-​out mice reveals on-​target Acad. Sci. USA 111, 18369–18374 (2014).
Bartholow, B. D. A cis-​eQTL in OPRM1 is associated activation maps in response to oxycodone. Front. 172. Galadrin, S. et al. The evasive nature of drug efficacy:
with subjective response to alcohol and alcohol use. Neurosci. 10, 471 (2016). implications for drug discovery. Trends Pharm. Sci. 28,
Alcohol Clin. Exp. Res. 41, 929–938 (2017). 150. Kenakin, T. The effective application of biased 423–430 (2007).
127. Smith, A. H. et al. Genome-​wide association study of signaling to new drug discovery. Mol. Pharmacol. 88, 173. Roth, B. L. et al. Salvinorin A: a potent naturally
therapeutic opioid dosing identifies a novel locus 1055–1061 (2015). occurring nonnitrogenous kappa opioid selective
upstream of OPRM1. Mol. Psychiatry 22, 346–352 151. Madariaga-​Mazon, A. et al. Mu-​opioid receptor biased agonist. Proc. Natl Acad. Sci. USA 99, 11934–11939
(2017). ligands: a safer and painless discovery of analgesics? (2002).
128. Mague, S. D. et al. Mouse model of OPRM1 (A118G) Drug Discov. Today 22, 1719–1729 (2017). 174. Sheffler, D. J. & Roth, B. L. Salvinorin A: the “magic
polymorphism has sex-​specific effects on drug-​ 152. Spangler, S. & Bruchas, M. R. Tuning biased GPCR mint” hallucinogen finds a molecular target in the
mediated behavior. Proc. Natl Acad. Sci. USA 106, signaling for physiological gain. Cell 171, 989–991 kappa opioid receptor. Trends Pharmacol. Sci. 24,
10847–10852 (2009). (2017). 107–109 (2003).
129. Ramchandani, V. A. et al. A genetic determinant of the 153. Kieffer, B. L. Drug discovery: designing the ideal 175. Cruz, A., Domingos, S., Gallardo, E. & Martinho, A. A
striatal dopamine response to alcohol in men. Mol. opioid. Nature 537, 170–171 (2016). unique natural selective kappa-​opioid receptor
Psychiatry 16, 809–817 (2011). 154. Bohn, L. M. et al. Enhanced morphine analgesia in mice agonist, salvinorin A, and its roles in human
This is a translational study to elucidate the role of lacking β-​arrestin 2. Science 286, 2495–2498 (1999). therapeutics. Phytochemistry 137, 9–14 (2017).
the MOR A118G SNP in response to alcohol, and 155. Raehal, K. M., Walker, J. K. & Bohn, L. M. Morphine 176. Butelman, E. R. & Kreek, M. J. Salvinorin A, a kappa-​
data show that 118G carriers in both humans and a side effects in β-​arrestin 2 knockout mice. J. Pharmacol. opioid receptor agonist hallucinogen: pharmacology
humanized A118G genetic mouse mutant release Exp. Ther. 314, 1195–1201 (2005). and potential template for novel pharmacotherapeutic
higher levels of DA following alcohol exposure. 156. DeWire, S. M. et al. A G protein-​biased ligand at the agents in neuropsychiatric disorders. Front. Pharmacol.
130. Bernardi, R. E. et al. A gene-​by-sex interaction for mu-​opioid receptor is potently analgesic with reduced 6, 190 (2015).
nicotine reward: evidence from humanized mice and gastrointestinal and respiratory dysfunction compared 177. Civelli, O. et al. G protein-​coupled receptor
epidemiology. Transl Psychiatry 6, e861 (2016). with morphine. J. Pharmacol. Exp. Ther. 344, deorphanizations. Annu. Rev. Pharmacol. Toxicol. 53,
131. Cabrera, E. A. et al. Neuroimaging the effectiveness of 708–717 (2013). 127–146 (2013).
substance use disorder treatments. J. Neuroimmune 157. Siuda, E. R., Carr, R. 3rd, Rominger, D. H. & Violin, J. D. 178. Reinscheid, R. K. et al. Orphanin FQ: a neuropeptide
Pharmacol. 11, 408–433 (2016). Biased mu-​opioid receptor ligands: a promising new that activates an opioidlike G protein-​coupled
132. Henriksen, G. & Willoch, F. Imaging of opioid receptors generation of pain therapeutics. Curr. Opin. Pharmacol. receptor. Science 270, 792–794 (1995).
in the central nervous system. Brain 131, 1171–1196 32, 77–84 (2017). 179. Meunier, J. C. et al. Isolation and structure of the
(2008). 158. Varadi, A. et al. Mitragynine/corynantheidine endogenous agonist of opioid receptor-​like ORL1
133. Kuwabara, H. et al. Mu opioid receptor binding pseudoindoxyls as opioid analgesics with mu agonism receptor. Nature 377, 532–535 (1995).
correlates with nicotine dependence and reward in and delta antagonism, which do not recruit 180. Zaveri, N. T. Nociceptin opioid receptor (NOP) as a
smokers. PLoS ONE 9, e113694 (2014). β-arrestin-2. J. Med. Chem. 59, 8381–8397 (2016). therapeutic target: progress in translation from
134. Nuechterlein, E. B., Ni, L., Domino, E. F. & Zubieta, J. K. 159. Schmid, C. L. et al. Bias factor and therapeutic window preclinical research to clinical utility. J. Med. Chem.
Nicotine-​specific and non-​specific effects of cigarette correlate to predict safer opioid analgesics. Cell 171, 59, 7011–7028 (2016).
smoking on endogenous opioid mechanisms. Prog. 1165–1175 (2017). 181. Toll, L., Bruchas, M. R., Calo, G., Cox, B. M. &
Neuropsychopharmacol. Biol. Psychiatry 69, 69–77 This work provides compelling evidence for the Zaveri, N. T. Nociceptin/orphanin FQ receptor
(2016). physiological relevance of MOR biased signalling, structure, signaling, ligands, functions, and interactions
135. Hermann, D. et al. Low mu-​opioid receptor status in as comparing biased signalling with behavioural with opioid systems. Pharmacol. Rev. 68, 419–457
alcohol dependence identified by combined positron effects of a range of MOR agonists reveals strong (2016).
emission tomography and post-​mortem brain analysis. correlation between Gi/β-​arrestin bias factors and 182. Witkin, J. M. et al. The biology of nociceptin/orphanin
Neuropsychopharmacology 42, 606–614 (2017). the analgesia–respiratory depression therapeutic FQ (N/OFQ) related to obesity, stress, anxiety, mood,
136. Mick, I. et al. Blunted endogenous opioid release window. and drug dependence. Pharmacol. Ther. 141,
following an oral amphetamine challenge in 160. Altarifi, A. A. et al. Effects of acute and repeated 283–299 (2014).
pathological gamblers. Neuropsychopharmacology treatment with the biased mu opioid receptor agonist 183. Sounier, R. et al. Propagation of conformational
41, 1742–1750 (2016). TRV130 (oliceridine) on measures of antinociception, changes during mu-​opioid receptor activation. Nature
137. Majuri, J. et al. Dopamine and opioid neurotransmission gastrointestinal function, and abuse liability in 524, 375–378 (2015).
in behavioral addictions: a comparative PET study in rodents. J. Psychopharmacol 31, 730–739 (2017). 184. Raehal, K. M., Schmid, C. L., Groer, C. E. & Bohn, L. M.
pathological gambling and binge eating. 161. Irannejad, R. et al. Functional selectivity of GPCR-​ Functional selectivity at the mu-​opioid receptor:
Neuropsychopharmacology 42, 1169–1177 (2017). directed drug action through location bias. Nat. Chem. implications for understanding opioid analgesia and
138. Karlsson, H. K. et al. Obesity is associated with Biol. 13, 799–806 (2017). tolerance. Pharmacol. Rev. 63, 1001–1019 (2011).
decreased μ-​opioid but unaltered dopamine D2 162. Urs, N. M. et al. Distinct cortical and striatal actions of 185. Pradhan, A. A., Smith, M. L., Kieffer, B. L. & Evans, C. J.
receptor availability in the brain. J. Neurosci. 35, a β-​arrestin-biased dopamine D2 receptor ligand Ligand-​directed signalling within the opioid receptor
3959–3965 (2015). reveal unique antipsychotic-​like properties. Proc. Natl family. Br. J. Pharmacol. 167, 960–969 (2012).
139. Saanijoki, T. et al. Opioid release after high-​intensity Acad. Sci. USA 113, E8178–E8186 (2016). 186. Ehrich, J. M. et al. Kappa opioid receptor-​induced
interval training in healthy human subjects. 163. Olson, K. M., Lei, W., Keresztes, A., LaVigne, J. & aversion requires p38 MAPK activation in VTA dopamine
Neuropsychopharmacology 43, 246–254 (2018). Streicher, J. M. Novel molecular strategies and targets neurons. J. Neurosci. 35, 12917–12931 (2015).
140. Nummenmaa, L. et al. Social touch modulates for opioid drug discovery for the treatment of chronic 187. Scherrer, G. et al. Knockin mice expressing fluorescent
endogenous mu-​opioid system activity in humans. pain. Yale J. Biol. Med. 90, 97–110 (2017). delta-​opioid receptors uncover G protein-​coupled
Neuroimage 138, 242–247 (2016). 164. Pasternak, G. W. Opioids and their receptors: are we receptor dynamics in vivo. Proc. Natl Acad. Sci. USA
141. Manninen, S. et al. Social laughter triggers there yet? Neuropharmacology 76, 198–203 103, 9691–9696 (2006).
endogenous opioid release in humans. J. Neurosci. (2014). This study provides a unique knock-​in mouse
37, 6125–6131 (2017). 165. Majumdar, S. et al. Truncated G protein-​coupled mu mutant line, and is the first of a series to visualize a
142. Nummenmaa, L. & Tuominen, L. Opioid system and opioid receptor MOR-1 splice variants are targets for GPCR in vivo; it shows DOR subcellular localization
human emotions. Br. J. Pharmacol. https://doi.org/ highly potent opioid analgesics lacking side effects. and trafficking, opening the way to understand
10.1111/bph.13812 (2017). Proc. Natl Acad. Sci. USA 108, 19778–19783 tolerance mechanisms.
143. Borsook, D., Becerra, L. & Hargreaves, R. A role for (2011). 188. Pradhan, A. A. et al. Ligand-​directed trafficking of the
fMRI in optimizing CNS drug development. Nat. Rev. 166. Convertino, M. et al. Mu-​opioid receptor delta-​opioid receptor in vivo: two paths toward
Drug Discov. 5, 411–424 (2006). 6-transmembrane isoform: a potential therapeutic analgesic tolerance. J. Neurosci. 30, 16459–16468
144. Becerra, L., Harter, K., Gonzalez, R. G. & Borsook, D. target for new effective opioids. Prog. (2010).
Functional magnetic resonance imaging measures of Neuropsychopharmacol. Biol. Psychiatry 62, 61–67 189. Su, D. et al. One-​step generation of mice carrying a
the effects of morphine on central nervous system (2015). conditional allele together with an HA-​tag insertion
circuitry in opioid-​naive healthy volunteers. Anesth. 167. Huang, W. et al. Structural insights into micro-​opioid for the delta opioid receptor. Sci. Rep. 7, 44476
Analg. 103, 208–216 (2006). receptor activation. Nature 524, 315–321 (2015). (2017).
145. Becerra, L. et al. Parallel buprenorphine phMRI 168. Spahn, V. et al. A nontoxic pain killer designed by 190. Siuda, E. R. et al. Spatiotemporal control of opioid
responses in conscious rodents and healthy human modeling of pathological receptor conformations. signaling and behavior. Neuron 86, 923–935
subjects. J. Pharmacol. Exp. Ther. 345, 41–51 (2013). Science 355, 966–969 (2017). (2015).

NATuRe RevIewS | NEuROSCiEnCE volume 19 | AUGUST 2018 | 513


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

191. Marchant, N. J. et al. Behavioral and physiological 201. Hendershot, C. S., Claus, E. D. & Ramchandani, V. A. 211. Henderson-​Redmond, A. N. et al. Morphine-induced
effects of a novel kappa-​opioid receptor-​based Associations of OPRM1 A118G and alcohol sensitivity antinociception and reward in “humanized” mice
DREADD in rats. Neuropsychopharmacology 41, with intravenous alcohol self-​administration in young expressing the mu opioid receptor A118G
402–409 (2016). adults. Addict. Biol. 21, 125–135 (2016). polymorphism. Brain Res. Bull. 123, 5–12
192. Marchant, N. J. et al. Role of ventral subiculum in 202. Bart, G. et al. Increased attributable risk related to a (2016).
context-​induced relapse to alcohol seeking after functional mu-​opioid receptor gene polymorphism in 212. Bilbao, A. et al. A pharmacogenetic determinant of
punishment-​imposed abstinence. J. Neurosci. 36, association with alcohol dependence in central mu-​opioid receptor antagonist effects on alcohol
3281–3294 (2016). Sweden. Neuropsychopharmacology 30, 417–422 reward and consumption: evidence from humanized
193. Harris, J. A. et al. Anatomical characterization of Cre (2005). mice. Biol. Psychiatry 77, 850–858 (2015).
driver mice for neural circuit mapping and 203. Schinka, J. A. et al. A functional polymorphism within
manipulation. Front. Neural Circuits 8, 76 (2014). the mu-​opioid receptor gene and risk for abuse of Acknowledgements
194. Cai, X. et al. Generation of a KOR-​Cre knockin mouse alcohol and other substances. Mol. Psychiatry 7, The authors deeply thank the CNRS/INSERM/University of
strain to study cells involved in kappa opioid signaling. 224–228 (2002). Strasbourg (France), the US National Institutes of Health
Genesis 54, 29–37 (2016). 204. Town, T. et al. Association of a functional mu-​opioid (National Institute of Drug Abuse Grant 05010 and National
195. Kumar, D., Chakraborty, J. & Das, S. Epistatic effects receptor allele (+118A) with alcohol dependency. Institute on Alcohol Abuse and Alcoholism Grant 16658 to
between variants of kappa-​opioid receptor gene and Am. J. Med. Genet. 88, 458–461 (1999). B.L.K.), the Canada Fund for Innovation and the Canada
A118G of mu-​opioid receptor gene increase 205. Dlugos, A. M. et al. OPRM1 gene variants modulate Research Chairs (to B.L.K and E.D.), and the Bourgeois family
susceptibility to addiction in Indian population. amphetamine-​induced euphoria in humans. Genes (B.L.K. is the Bourgeois Chair for Pervasive Developmental
Prog. Neuropsychopharmacol. Biol. Psychiatry 36, Brain Behav. 10, 199–209 (2011). Disorders) for continuous support.
225–230 (2012). 206. Schuck, K., Otten, R., Engels, R. C. & Kleinjan, M.
196. Deb, I., Chakraborty, J., Gangopadhyay, P. K., Initial responses to the first dose of nicotine in novel Author contributions
Choudhury, S. R. & Das, S. Single-​nucleotide smokers: the role of exposure to environmental E.D. and B.L.K. researched data for the article, made substan-
polymorphism (A118G) in exon 1 of OPRM1 gene smoking and genetic predisposition. Psychol. Health tial contributions to the discussion of content, wrote the arti-
causes alteration in downstream signaling by mu-​ 29, 698–716 (2014). cle and reviewed and edited the manuscript before
opioid receptor and may contribute to the genetic risk 207. Zhang, Y. et al. Mouse model of the OPRM1 (A118G) submission.
for addiction. J. Neurochem. 112, 486–496 (2010). polymorphism: differential heroin self-​administration
197. Kapur, S., Sharad, S., Singh, R. A. & Gupta, A. K. A118G behavior compared with wild-​type mice. Competing interests
polymorphism in mu opioid receptor gene (OPRM1): Neuropsychopharmacology 40, 1091–1100 (2015). The authors declare no competing interests.
association with opiate addiction in subjects of Indian 208. Browne, C. A., Erickson, R. L., Blendy, J. A. & Lucki, I.
origin. J. Integr. Neurosci. 6, 511–522 (2007). Genetic variation in the behavioral effects of Publisher’s note
198. Bart, G. et al. Substantial attributable risk related to a buprenorphine in female mice derived from a murine Springer Nature remains neutral with regard to jurisdictional
functional mu-​opioid receptor gene polymorphism in model of the OPRM1 A118G polymorphism. claims in published maps and institutional affiliations.
association with heroin addiction in central Sweden. Neuropharmacology 117, 401–407 (2017).
Mol. Psychiatry 9, 547–549 (2004). 209. Briand, L. A. et al. Mouse model of OPRM1 (A118G) Reviewer information
199. Szeto, C. Y., Tang, N. L., Lee, D. T. & Stadlin, A. polymorphism increases sociability and dominance Nature Reviews Neuroscience thanks E. J. Nestler and the
Association between mu opioid receptor gene and confers resilience to social defeat. J. Neurosci. 35, other anonymous reviewer(s) for their contribution to
polymorphisms and Chinese heroin addicts. 3582–3590 (2015). the peer review of this work.
Neuroreport 12, 1103–1106 (2001). 210. Robinson, J. E. et al. Receptor reserve moderates
200. Li, T. et al. Association analysis of polymorphisms in mesolimbic responses to opioids in a humanized Supplementary information
the mu opioid gene and heroin abuse in Chinese mouse model of the OPRM1 A118G polymorphism. Supplementary information is available for this paper at
subjects. Addict. Biol. 5, 181–186 (2000). Neuropsychopharmacology 40, 2614–2622 (2015). https://doi.org/10.1038/s41583-018-0028-x.

514 | AUGUST 2018 | volume 19 www.nature.com/nrn


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like