Opioid-receptor-heteromer-specific trafficking and pharmacology

https://doi.org/10.1016/j.coph.2009.09.007Get rights and content

Homomerization and heteromerization of 7 transmembrane spanning (7TM)/G-protein-coupled receptors (GPCRs) have been an important field of study. Whereas initial studies were performed in artificial cell systems, recent publications are shifting the focus to the in vivo relevance of heteromerization. This is especially apparent for the field of opioid receptors. Drugs have been identified that selectively target opioid heteromers of the delta-opioid receptor with the kappa and the mu-opioid receptors that influence nociception and ethanol consumption, respectively. In addition, in several cases, the specific physiological response produced by the heteromer may be directly attributed to a difference in receptor trafficking properties of the heteromers compared with their homomeric counterparts. This review attempts to highlight some of the latest developments with regard to opioid receptor heteromer trafficking and pharmacology.

Introduction

Opioids, and the receptors to which they bind, have been extensively studied and are well known for their roles in pain modulation/analgesia and reward. The peptidergic opioid receptors belong to class A of the family of 7 transmembrane spanning (7TM)/G-protein-coupled receptors (GPCRs). For many years, opioid receptors were subdivided into three classes: the mu-opioid receptor (MOP-R), the delta-opioid receptor (DOP-R), and the kappa-opioid receptor (KOP-R), each of which displays at least two pharmacological ‘subtypes’ in vivo [1, 2, 3]. In the early 1990s sequence homology cloning efforts resulted in the identification of a fourth ‘opioid’ receptor, the nociceptin (orphanin FQ/ORL1) receptor NOP-R, which despite its high homology to the other opioid receptors does not bind ‘classic’ opioid ligands with high affinity [4]. Continuing research at the molecular level has resulted in the identification of receptor splice variants of each of these receptors [5]. An additional layer of complexity is added by the ability of opioid receptors to engage in receptor–receptor interactions, forming receptor homomers and heteromers with altered pharmacological properties [6, 7], first described more than a decade ago [8]. Hence, although only four opioid receptor genes have been identified, the pharmacological diversity in opioid response is much greater.

An important aspect of opioid pharmacology is the establishment of tolerance. Opioid tolerance can have multiple causes, but often include mechanisms involving receptor trafficking. These mechanisms, such as receptor phosphorylation/desensitization, internalization, recycling, or degradation, for a large part, rely on receptor interactions with non-7TM/GPCRs, such as protein kinases and β-arrestins [9, 10].

This review focuses on recent advances in understanding opioid receptor heteromerization and trafficking.

Not only do the opioid receptor subtypes vary in their signaling and ligand-binding properties, but, important, they also display different trafficking properties. The most studied receptors in this respect are the MOP-Rs, which recycle after ligand activation, and the DOP-Rs, which degrade after activation [11]. However, this is not as straightforward as it seems, as the MOP-R agonist morphine does not induce receptor internalization and/or recycling and there are endogenous ligands that both do and do not differentiate between MOP-R and DOP-R [4]. Both the ability and the inability of opioid receptors to desensitize and endocytose have been proposed as mechanisms responsible for opioid tolerance [12]. Nevertheless, it is clear that desensitization alone cannot explain morphine tolerance, because active receptors still remain in the ‘tolerant state’ and are revealed as signs of physical withdrawal on removal of a drug.

Importantly, several studies have suggested that enhancing MOP-R desensitization and endocytosis can delay tolerance [13••, 14••]. Indeed, endogenous endorphins [14••], and exogenous DAMGO [15] or methadone [16] all appear to enhance the ability of morphine to promote endocytosis of the MOP-R. One possible explanation for these findings is that morphine-occupied MOP-Rs are endocytosed because they form homomers with MOP-Rs occupied by an endocytosis-promoting ligand. Thus, these findings hint at the possible existence of MOP-R homomers [15] (Figure 1A).

In addition, recent studies propose that DOP-Rs are located predominantly intracellularly, but can be translocated to the cell surface under several conditions (for review see [17] and Figure 2A), for example chronic morphine exposure, stress, inflammation, or more recently, ethanol consumption [18]. It appears that both substance P [19] and the presence of MOP-Rs may play a role in the translocation of DOP-Rs to the surface [20, 21]. However, the redistribution of intracellular DOP-Rs remains controversial, with recent findings arguing against this hypothesis [22••].

7TM/GPCRs were initially thought to function solely as monomeric entities. However, a vast amount of data has been amassed over the last two decades suggesting that 7TM/GPCRs can interact with themselves or each other to form receptor homomers and heteromers [6]. The opioid receptors were some of the first heteromers that were comprehensively studied using functional (changes in pharmacology) as well as biochemical (immunoprecipitation, crosslinking) and biophysical (FRET and BRET) methods [23]. The DOP-R can form heteromers with the MOP-R [24, 25, 26, 27] as well as the KOP-R [27, 28]. The MOP-R can also heteromerize with the NOP-R [29]. The existence of MOP/KOP-R heteromers is uncertain; Wang et al. showed that all opioid receptors have a similar affinity to form receptor homomers and heteromers [27]. However, other studies found no such interactions [28].

It is possible that opioid heteromers could represent some of the opioid receptor subtypes that have been pharmacologically defined, but not attributed to splice variants, such as DOP-R1 (with a preference for DPDPE and BNTX) and DOP-R2 (with a preference for deltorphin II and naltriben) [3]. For example, there is some evidence that the DOP-R1 could be a DOP/KOP-R heteromer [30, 31], whereas the DOP-R2 could be a DOP/MOP-R heteromer [32]. However, Gomes et al. found that the DOP/MOP-R heteromer showed enhanced affinity not only for some DOP-R1 (BNTX) but also for some DOP-R2 (deltorphin II) ligands but not for others (DPDPE and naltriben) [33]. Intriguingly, some behavioral effects of DOP-R1, but not DOP-R2 ligands, are affected by disruption of MOP-R, again suggesting that the DOP-R1 is a DOP/MOP-R heteromer [34••, 35].

Similar to the DOP-R, two MOP-R subtypes exist: MOP-R1, which is inhibited by naloxonazine, and MOP-R2 which is not [1]. Three KOP-R subtypes have thus far been identified pharmacologically: KOP-R1 binds arylacetamides and KOP-R2 does not [2], whereas KOP-R3 [36] is insensitive to the KOP-R ligand U50,488. Some of these subtypes may actually represent the same receptors, for example MOP-R2 may be DOP-R2 [37], whereas DOP-R1 and KOP-R2 may be the same DOP/KOP-R heteromer [30]. In addition, opioid receptors may engage in the formation of heteromers with receptors outside the opioid receptor family. MOP-R has been described to form heteromers with many receptors including NK1 [38], CCR5 [39], and the α2A-adrenergic receptors [40], whereas both DOP-R and KOP-R can form receptor heteromers with the β2-adrenergic receptor [41]. Importantly, these heteromers typically exhibit unique pharmacological properties, showing differences in ligand affinities, signaling, and receptor trafficking.

It is difficult to distinguish whether the need for two opioid receptors to produce a biological effect is due to heteromerization or convergence in a circuit. Importantly, in at least one case, the MOP-R1 and DOP-R1 have been shown to coimmunoprecipitate in CNS tissues [25] and antibodies that selectively recognize the DOP/MOP-R heteromer in vitro recognize a receptor complex in vivo as well [42]. Furthermore, in at least one case, a ligand has been identified that selectively activates a heteromer in vitro and produces a biological effect in vivo [43]. Thus, although there is still some debate as to the in vivo role of DOP/MOP-R or DOP/KOP-R heteromers, there is mounting evidence that these targets do exist, at least in some tissues, in vivo.

There is still much ongoing debate on the ontogeny and fate of receptor heteromers. It is yet to be verified whether receptor homomers and heteromers are formed firstly, in the endoplasmatic reticulum and Golgi apparatus during receptor synthesis and maturation or secondly, while expressed on the cell surface. As an evidence of this debate, early studies suggested that the heteromerization of DOP/MOP-R might only occur at the cell surface [26], whereas other reports have shown that DOP/MOP-R heteromers are formed in the ER [44••]. In addition, a group of receptor chaperone proteins, called receptor transporting proteins (RTPs) and receptor expression enhancing proteins (REEPs), have recently been identified, which may assist in the transport of heteromers to the cell surface [42•, 45] (Figure 2A).

Following their endocytosis, the MOP-R and DOP-R appear to be sorted to different compartments. Specifically, while the MOP-R is recycled to the plasma membrane [46, 47], the DOP-R is targeted for degradation [11]. Thus, from a postendocytic trafficking viewpoint, the DOP/MOP-R heteromer could display a unique trafficking profile, although there is some question as to whether the DOP/MOP-R internalizes as receptor heteromer in vitro [26]. In addition, heteromerization of the KOP-R and DOP-R also appears to affect trafficking. For example, while DOP-Rs are rapidly endocytosed in response to activation by etorphine, KOP-Rs are not, and when the two receptors are coexpressed KOP-Rs prevent the endocytosis of DOP-Rs [48].

Heteromers between the β2-AR and either the DOP-R or the KOP-R also display altered trafficking properties compared with receptor homomers/monomers. For example, when the β2-AR and DOP-R are coexpressed, activation of either receptor leads to cointernalization of the other receptor. On the other hand, the β2-AR/KOP-R heteromer does not endocytose when activated by agonists for either protomer [41] (Figure 2B). Similarly, when the MOP-R and NK1 receptors [38] are coexpressed, the receptors cointernalize with activation by agonists selective for either protomer. Heteromerization between MOP-R and NK1-R could explain why there is a loss of morphine place preference in NK1−/− mice [49] and why chronic morphine treatment alters substance P-induced internalization of the NK1-R [50]. On the other hand, MOP-R heteromerization with CCR5 only produces crossdesensitization, but not crossinternalization [39].

A recent finding suggests that MOP-Rs are located in lipid rafts on the cell surface, but when activated by specific ligands (etorphine, but not morphine) move out of these rafts [51]. Mechanisms similar to this could play a role in the formation or breakup of heteromers and shed more light on the receptor heteromer ontogeny (Figure 2C).

One method of targeting homomers and heteromers is the coadministration of two drugs that can interact with the two binding pockets of the homomer/heteromer (Figure 1A). For instance, it was suggested that the DOP-R antagonist TIPPψ enhances morphine analgesia in vivo, by acting on the DOP/MOP-R heteromer [25]. In addition, several efforts have been made to design single ligands that bridge opioid heteromers and, thus, potentially bring together individual receptors into heteromeric complexes and/or stabilize an existing heteromer. The early work in this area targeted homomers and was motivated by a desire to develop both tools to distinguish between different types of opioid receptors and more potent ligands. These ligands often did exhibit enhanced potency (presumably caused by their higher affinity) [52]. Recently, these efforts have been expanded to probe the existence of heteromers, receptor allosteric coupling, and potential novel antinociceptive sites [53, 54, 55]. For example, the mu–delta agonist–antagonist (MDAN) series of ligands consist of a MOP-R agonist (oxymorphone) and a DOP-R antagonist (naltrindole) moiety linked by a spacer of variable length. The underlying hypothesis behind these ligands is the finding that DOP-R anatagonists (naltrindole) attenuate acute morphine tolerance and dependence [56]. Some of the ligands from the MDAN series show activities in vivo consistent with a unique activity at a heteromer, that is reduced tolerance and naloxone precipitated withdrawal signs. This is exemplified by the finding that the novel properties of one molecule in this series, MDAN-19, cannot be recapitulated by adding the two pharmacophores separately, and that its novel properties are dependent on the spacer length (Figure 1B). The proposed mechanism for these bivalent ligands is the ability of the DOP-R antagonist to negatively modulate the MOP-R receptor [54]. In addition, the bivalent ligand KDN-21 is a DOP/KOP-R bivalent antagonist that blocks antinociception produced by both the DOP-R1 agonist DPDPE and the KOP-R2 agonist bremazocine in the spinal cord [30]. Several bivalent ligands have been synthesized that have dual affinity for MOP-R and KOP-R [57, 58] as well as ‘tri-functional’ ligands harboring DOP-R antagonistic, MOP-R agonistic, and KOP-R partial agonistic properties [59]. Some of these ligands have recently been tested in vivo [60]. However, although these ligands were potent analgesics, activity did not differ from the monovalent parent compounds, and therefore, are not uniquely targeting KOP/MOP-R heteromers. In short, as all bivalent ligands described to date retain activity at homomers/monomers, it has been difficult to draw conclusions as to the existence or functional importance of heteromers using these tools.

However, there is at least one case in which a ligand has been described that shows novel activity at an opioid heteromer distinct from that on either homomer, and is an analgesic in vivo. 6′-GNTI is a small molecule with high affinity for DOP-R and KOP-R. However, although this ligand is an antagonist at DOP-R and a weak partial agonist at KOP-R, it is a potent agonist on DOP/KOP-R heteromers and produces antinociception when administered intrathecally [43], consistent with the localization of DOP/KOP-R heteromers in the spinal cord, but not in the brain [43] (Figure 1C).

The availability of ligands with selective or novel activity at receptor heteromers would enable not only the study of heteromer localization in vivo, but also their dynamics as well. In addition, if opioid receptor heteromers are expressed in a tissue-selective manner, they could be exploited to prevent the side effects of opiates, such as respiratory depression, constipation, and dependence that arise as a consequence of systemic use of these drugs. In addition, if the expression of select heteromers is altered during the development of morphine tolerance, they could represent unexplored and selective targets for reversing, preventing or modulating tolerance, and dependence. Furthermore, receptor heteromers could be expressed in a gender-specific manner, and thereby explain some of the gender-specific effects of opioids, in particular, KOP-R opioids [61].

By way of an example, 6′-GNTI was an effective analgesic only when administered in the spinal cord, but not when injected intracerebroventricularly [43]. Thus, a drug similar to 6′-GNTI could show reduced side effects such as respiratory depression if the target of 6′-GNTI (the DOP/KOP-R heteromer) is selectively expressed in the spinal cord. In another example, TAN-67, a DOP-R1 selective agonist, reduces ethanol consumption in mice. The activity of TAN-67 is dependent on the presence of DOP-R as well as MOP-R (Figure 1B). These results suggest that DOP-R1 is a DOP/MOP-R heteromer that can be selectively targeted with an agonist to reduce ethanol consumption [34••], without the side effects of disphoria produced by opioid receptor antagonists.

In addition, drug ‘cocktails’ that target either homomer or heteromers could be therapeutically valuable. For example, an opioid cocktail consisting of morphine and either methadone or DAMGO promotes morphine-induced endocytosis, and thus, takes advantage of the homomeric nature of the MOP-R, leading to reduced tolerance and dependence [15, 16•], Figure 1A. Another example is a cocktail of morphine and serotonin, which drives the endocytosis of the MOP-R/5-HT2A heteromer, suggesting that serotonin might be expected to reduce the development of tolerance and dependence to morphine also by altering receptor trafficking [62].

Section snippets

Conclusions

Although evidence in support of the existence of opioid receptor homomers and heteromers continues to accumulate, their functional relevance remains ambiguous [63]. In most studies to date, receptor heteromers have been either recombinantly coexpressed in a cell line or studied in cell lines that endogenously express receptors at physiological levels. However, utilizing these systems for the detection of novel activities at heteromers is challenging at best, because in all cases both homomers

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

This work was supported by grants from the Austrian Science Fund (P18723), the Jubiläumsfonds of the Austrian National Bank and the Lanyar Stiftung Graz (all to MW). JLW and RvR were supported by funds provided by the State of California for medical research through the University of California San Francisco, by the NIH National Institute on Drug Abuse Grants DA015232 and DA019958 and DOD Grant W81XWH-08-1-0005.

References (66)

  • B.A. Jordan et al.

    G-protein-coupled receptor heterodimerization modulates receptor function

    Nature

    (1999)
  • Y. Pan et al.

    Dimerization of morphine and orphanin FQ/nociceptin receptors: generation of a novel opioid receptor subtype

    Biochem Biophys Res Commun

    (2002)
  • P.S. Portoghese et al.

    Identity of the putative delta1-opioid receptor as a delta–kappa heteromer in the mouse spinal cord

    Eur J Pharmacol

    (2003)
  • R.M. Van Rijn et al.

    The delta-1 opioid receptor is a heterodimer that opposes the actions of the delta-2 receptor on alcohol intake

    Biol Psychiatry

    (2009)
  • J.A. Clark et al.

    Kappa opiate receptor multiplicity: evidence for two U50,488-sensitive kappa 1 subtypes and a novel kappa 3 subtype

    J Pharmacol Exp Ther

    (1989)
  • C. Chen et al.

    Heterodimerization and cross-desensitization between the micro-opioid receptor and the chemokine CCR5 receptor

    Eur J Pharmacol

    (2004)
  • B.A. Jordan et al.

    Oligomerization of opioid receptors with beta 2-adrenergic receptors: a role in trafficking and mitogen-activated protein kinase activation

    Proc Natl Acad Sci U S A

    (2001)
  • A. Hasbi et al.

    Trafficking of preassembled opioid mu–delta heterooligomer-Gz signaling complexes to the plasma membrane: coregulation by agonists

    Biochemistry

    (2007)
  • R.M. Van Rijn et al.

    The only way is up: preventing opioid tolerance by promoting cell surface expression of MOR–DOR heterodimers?

    Mol Interv

    (2008)
  • E.E. Abdelhamid et al.

    Selective blockage of delta opioid receptors prevents the development of morphine tolerance and dependence in mice

    J Pharmacol Exp Ther

    (1991)
  • J.L. Neumeyer et al.

    Design and synthesis of novel dimeric morphinan ligands for kappa and micro opioid receptors

    J Med Chem

    (2003)
  • R.S. Zukin et al.

    Characterization and visualization of rat and guinea pig brain kappa opioid receptors: evidence for kappa 1 and kappa 2 opioid receptors

    Proc Natl Acad Sci U S A

    (1988)
  • P.A. Zaki et al.

    Opioid receptor types and subtypes: the delta receptor as a model

    Annu Rev Pharmacol Toxicol

    (1996)
  • M. Waldhoer et al.

    Opioid receptors

    Annu Rev Biochem

    (2004)
  • S. Ferre et al.

    Building a new conceptual framework for receptor heteromers

    Nat Chem Biol

    (2009)
  • R.R. Gainetdinov et al.

    Desensitization of G protein-coupled receptors and neuronal functions

    Annu Rev Neurosci

    (2004)
  • J.L. Whistler et al.

    Modulation of postendocytic sorting of G protein-coupled receptors

    Science

    (2002)
  • J.T. Williams et al.

    Cellular and synaptic adaptations mediating opioid dependence

    Physiol Rev

    (2001)
  • C. Zollner et al.

    Chronic morphine use does not induce peripheral tolerance in a rat model of inflammatory pain

    J Clin Invest

    (2008)
  • L. He et al.

    Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization

    Cell

    (2002)
  • L. He et al.

    An opiate cocktail that reduces morphine tolerance and dependence

    Curr Biol

    (2005)
  • J.-S. Guan et al.

    Interaction with vesicle luminal protachykinin regulates surface expression of d-opioid receptors and opioid analgesia

    Cell

    (2005)
  • C.M. Cahill et al.

    Prolonged morphine treatment targets delta opioid receptors to neuronal plasma membranes and enhances delta-mediated antinociception

    J Neurosci

    (2001)
  • Cited by (89)

    • Transient nature of stable behavioural patterns, and how we can respect it

      2022, Current Opinion in Behavioral Sciences
      Citation Excerpt :

      It proceeds in several stages, and each stage involves a cascade of contingent transformations regulated by mediators such as GABA, Glutamate, G-protein coupled receptors, transcription and neurotrophic factors, enzymes, metabolites, ATP, calcium and other chemical systems, as well as the regulatory impact of NTs and NPs on each other [6••]. Similar complexity and contingent constructions were described in the action of opioid receptors (OR) [7••,8]. Therefore (and third), the success of each of these multiple stages is far from being guaranteed and depends not only on the availability of mediators but also on the physiological state of the body, of the supporting microglia cells, of the brain cells that manufacture the needed components, and of environmental factors.

    • New Insights into Modes of GPCR Activation

      2018, Trends in Pharmacological Sciences
    View all citing articles on Scopus
    View full text