Abstract
In recent years, the delta opioid receptor has attracted Increasing Interest as a target for the treatment of chronic pain and emotional disorders. Due to their therapeutic potential, numerous tools have been developed to study the delta opioid receptor from both a molecular and a functional perspective. This review summarizes the most commonly available tools, with an emphasis on their use and limitations. Here, we describe (1) the cell-based assays used to study the delta opioid receptor. (2) The features of several delta opioid receptor ligands, including peptide and non-peptide drugs. (3) The existing approaches to detect delta opioid receptors in fixed tissue, and debates that surround these techniques. (4) Behavioral assays used to study the in vivo effects of delta opioid receptor agonists; including locomotor stimulation and convulsions that are induced by some ligands, but not others. (5) The characterization of genetically modified mice used specifically to study the delta opioid receptor. Overall, this review aims to provide a guideline for the use of these tools with the final goal of increasing our understanding of delta opioid receptor physiology.
Keywords: pain, cell lines, mutant mice, G protein-coupled receptor
INTRODUCTION
Opioids have been used for centuries, during which time they have been alternately hailed as both a panacea of man’s ills, and cursed as a scourge of civilization. The vast majority of opioids used clinically (eg. morphine, hydrocodone, oxycodone) or recreationally (eg. heroin) produce their behavioral effects primarily through activation of the mu opioid receptor. Although mu agonists are incredibly potent analgesics, they also produce deleterious adverse effects which severely limit their therapeutic use. Activation of the mu opioid receptor can produce constipation, respiratory depression, and sedation. Furthermore, tolerance and 1 physical dependence is observed after chronic use, and the abuse liability of mu agonists is very high.
Another member of the opioid receptor family - the delta opioid receptor – may offer a potential alternative to mu-based therapies. In acute pain states, activation of the delta opioid receptor is relatively ineffective compared to their mu counterparts (Gallantine and Meert, 2005). However, they are highly effective in chronic pain states (Hurley and Hammond, 2000; Fraser et al., 2000a; Cahill et al., 2003; Nadal et al., 2006; Gaveriaux-Ruff et al., 2008; Pradhan et al., 2009, 2010), and delta agonists were also recently shown to be effective for the treatment of migraine (Pradhan et al., 2014). Further, activation of the delta opioid receptor appears to have low abuse liability, as delta agonists are not self-administered (Negus et al., 1998; Stevenson et al., 2005) nor do they cause physical dependence (Brandt et al., 2001). Interestingly, delta opioid receptors also positively modulate emotional tone. Genetic deletion of either the delta opioid receptor or its endogenous ligand, enke-phalin, results in anxiogenic and depressive-like behaviors (Konig et al., 1996; Filliol et al., 2000). In contrast, delta agonists can produce anxiolytic and anti-depressant effects (Saitoh et al., 2004; Perrine et al., 2006). This emotional regulation may be particularly important for the treatment of chronic pain states, as there is a high comorbidity with depression and anxiety (Yalcin and Barrot, 2014). The potential of targeting this receptor has not gone unnoticed, and there are ongoing efforts to develop delta opioid receptor agonists for the treatment of a number of psychiatric and neurological conditions (Pradhan et al., 2011a).
During the past 5 years, there have been many reviews published on the delta opioid receptor, which have highlighted their biological function, therapeutic utility, and signaling mechanisms; and we direct the reader to these excellent resources (Pradhan et al., 2011a; Chu Sin Chung and Kieffer, 2013; van Rijn et al., 2013; Gendron et al., 2014; Charfi et al., 2015; Klenowski et al., 2015). The aim of this review, however, is to provide a more practical guide on how to study the delta opioid receptor. Our intention is to discuss the tools that are available to probe delta opioid receptor signaling, distribution, and function; and to highlight some of the pros and cons associated with each method. We hope that this review will be a useful “how to” guide for those interested in working on the delta opioid receptor.
How to examine delta opioid receptors in cells
The expression of delta opioid receptors in non-neuronal cell lines has served as a powerful tool to delineate delta opioid receptor trafficking and regulation (Whistler et al., 2002; Puthenveedu et al., 2010; Henry et al., 2011), signaling complexes (Audet et al., 2008, 2012; Charfi et al., 2014), and a multitude of other mechanisms. Delta opioid receptors have been transfected into several non-neuronal cell lines, such as the Chinese Hamster Ovary (CHO), the Human Embryonic Kidney (HEK) 293, and the African Green Monkey Kidney (COS-7) (Chan et al., 2003; Hong et al., 2009; Tudashki et al., 2014; Nagi et al., 2015a). Cells are transfected with cDNA encoding the delta opioid receptor, modified to express a peptide epitope or tag within either the N-terminal extracellular region or the C-terminus. Widely used epitope tags include hemagglutinin (HA), c-myc, and FLAG (Qiu et al., 2007; Tudashki et al., 2014; Nagi et al., 2015a). Epitope tagging facilitates the detection of the receptor by high-affinity antibodies, and overcomes many of the difficulties posed by using delta opioid receptor specific antibodies (see section below). In addition, delta opioid receptors can also be modified for FRET/BRET- (fluores-cence/bioluminescence resonance energy transfer) based technologies, thus allowing for the direct visualization of specific protein-protein interactions (Audet et al., 2008, 2012; Richard-Lalonde et al., 2013; Charfi et al., 2014; Nagi et al., 2015b; Pradhan et al., 2016).
However, it is important to keep in mind some of the limitations of transfected cell systems. There is the possibility that tagging the receptor may affect receptor function. For example, tags can interfere with post-translational processing (Jiang et al., 2012), oligomerization, ligand binding (N-terminal tags), and receptor regulatory and signaling events (C terminus). In addition, some tags may also affect receptor distribution within the cell itself ((Wang et al., 2008) and see detection section below). Furthermore, each cell line will express different types and levels of G proteins and regulatory proteins that will influence the outcome of the study. For example, delta opioid receptor internalization was found to be regulated by different proteins in rat primary neuronal cultures versus HEK 293 cells (Charfi et al., 2014). Also, microarray analysis has revealed several differences in gene expression of GPCRs and different signaling molecules between HEK 293 cells, AtT20, BV2 and N18 cell lines (Atwood et al., 2011). Moreover, transfection can often result in overexpression of the receptor, which also alters receptor regulation and function. Expression level may also vary across passages or batches of cells and across labs.
As an alternative to recombinant cells, there are neuronal lines that endogenously express the delta opioid receptor that may be considered closer to an endogenous environment. However, the lack of a tag means that receptor detection is more restricted. A further caveat of the use of immortalized hybrid cell lines is that they tend to lose chromosomes as they divide (Heumann et al., 1977; Geraghty et al., 2014). Therefore, it is recommended to create a large stock of frozen cells of low passage number, strictly track the number of passages, use the hybrid cells in a defined bracket of passage numbers, and report which passages were used for publication. The genetic instability of these cells lines can lead to batch variability and could contribute to variations in results between different laboratories. Some of the most commonly used cells lines that endogenously express the delta opioid receptor are the following:
NG108-15 cell line
These cells were obtained by inactivated Sendai virus-induced fusion of mouse | N18TG2 neuroblastoma cells with rat C6-BU-1 glioma cells (Klee and Nirenberg, 1974). They served as a source for the initial cloning of the delta opioid receptor (Evans et al., 1992; Kieffer et al., 1992), and have long been used as a cellular model for this receptor (Law et al., 1982, 1983; Hsia et al., 1984; Moses and Snell, 1984; Cone et al., 1991; Roerig et al., 1992). They carry a homogenous population of endogenous delta opioid receptor, and do not express mu or kappa opioid receptors (Chang and Cuatrecasas, 1979). However, they do express the opioid receptor-like receptor (ORL1 (Ma et al., 1997)). Importantly, NG108-15 cells rely to a large extent on anaerobic glycolysis for energy production. This gives rise to high concentrations of lactic acid, producing cell dissociation and loss of viability (Hamprecht et al., 1985). A further important issue to keep in mind is the instability of delta opioid receptor expression in this line; where early passages have higher probability of expressing the delta opioid receptor (passage number <30).
F11
This is a hybrid cell line created from the fusion of embryonic rat dorsal root ganglia (DRG) neurons with cells from mouse neuroblastoma N18TG2 (Platika et al., 1985). These cells exhibit properties characteristic of DRG neurons including neuronal morphology, expression of cell-surface gangliosides and excitable membranes (Platika et al., 1985). They express mu and delta opioid receptors that inhibit adenylate cyclase through pertussis toxin-sensitive Gi/o proteins (Francel et al., 1987; Fan et al., 1992). Overall, F11 cells are a useful tool to study delta opioid receptors in a nociceptor-like cell model that displays properties similar to sensory DRGs.
SK-N-BE cell line
This is a human neuroblastoma cell line established from a bone marrow biopsy, which endogenously expresses the human delta opioid receptor (Polastron et al., 1994). It has been proposed that this cell line expresses two delta opioid receptor subtypes, δ1 and δ2 (Polastron et al., 1994; Allouche et al., 2000). However, there is still controversy on the existence of delta opioid receptor subtypes since the different abilities of delta agonists to stimulate the receptor can also be explained by other means (different ligand-binding sites at the same receptor, biased-agonism, heterodimers etc). SK-N-BE cells may also express mu opioid receptors at a similar ratio to delta (Palazzi et al., 1996). These cells have been used extensively to study ligand directed signaling at the delta opioid receptor (Allouche et al., 1999a,b; Marie et al., 2003b, 2008).
SK-N-SH cell line
This is a human neuroblastoma cell line established from a bone marrow biopsy, which contains both mu and delta opioid receptors in a µ/δ ratio of ~5:1 (Yu et al., 1986). However, phenotypic heterogeneity in this range has been described due to differences in culture conditions (Baumhaker et al., 1993). This cell line has been used extensively to study interactions between the mu and delta opioid receptor (Baumhaker et al., 1993), and the formation of mu-delta. heterodimers specifically (Gomes et al., 2000, 2004). Work with this cell line has also revealed a stimulatory effect of opioid peptides on adenylate cyclase (Same et al., 1998; Rubovitch et al., 2003). In addition to mu and delta opioid receptors, SK-N-SH cells also express ORL-1 receptors (Cheng et al., 1997).
SH-SY5Y cell line
This cell line was subcloned from the SK-N-SH line, and express mu and delta opioid receptors (Kazmi and Mishra, 1986; Yu and Sadee, 1988). Like its parent line, SH-SY5Y cells express delta opioid receptors to a lesser extent than mu (Kazmi and Mishra, 1987; Yu and Sadee, 1988). SH-SY5Y cells also express ORL-1 receptors (Cheng et al., 1995), and signaling proteins like the Regulator of G protein signaling 4 (RGS4) (Lambert et al., 1989; McDonald et al., 1994; Wang et al., 2009; Levitt et al., 2011; Wang and Traynor, 2011). Given its repertoire of signaling proteins, SH-SY5Y cells have been widely used for the study of mu and delta opioid receptor activity, and receptor desensitization (Elliott et al., 1997; Nowoczyn et al., 2013). In addition, it has been reported that mu and delta opioid receptors share a common pool of G proteins and adenylate cyclase in SH-SY5Y cells (Alt et al., 2002; Levitt et al., 2011), indicating the close proximity of both receptors in these cells; however, no direct evidences of mu-delta heterodimerization have been found in this cell line. In addition, when these cells are used in a differentiated state they show a more mature neuronal phenotype and upregulated expression of opioid receptors (Yu and Sadee, 1988).
What ligands bind to the delta opioid receptor
There are many ligands that bind to the delta opioid receptor, and the aim of this section is to cover only those that are most commonly used and commercially available (Table 1). A wide range of peptide and small molecules have been developed that target the delta opioid receptor. Among the peptide delta opioid receptor agonists, the most popular are [d-Ala2, d-Leu5]-enkephalin (DADLE), [d-Pen2,5]-enkephalin (DPDPE), deltorphin I and deltorphin II. DADLE and DPDPE were obtained through chemical modification of the endogenous peptides met- and leu-enkephalin (Beddell et al., 1977; Mosberg et al., 1983). DADLE and DPDPE possess higher delta opioid receptor selectivity and stability than their prototypes, although DADLE appears to have less selectivity to the delta opioid receptor (Goldstein and Naidu, 1989), and both agonists have affinity for the mu opioid receptor as well (Fraser et al., 2000b; Chan et al., 2003). Deltorphin I and II were isolated from frog skin extracts (Erspamer et al., 1989). In comparison with the enkephalin analogs (DADLE and DPDPE), deltorphin I and II have higher biostability and delta opioid receptor selectivity, and comparatively deltorphin II has greater selectivity for the delta opioid receptor over mu (Payza, 2004). Peptide antagonists have also been developed against the delta opioid receptor, specifically TIPP, TIPP-NH2 and its analogs (Schiller et al., 1992a,b). Among them, TIPP[ψ] has shown subnanomolar affinity and extraordinary selectivity for the delta opioid receptor (Schiller et al., 1993).
Table 1.
Peptide and non-peptide delta opioid receptor ligands
Although peptide agonists have been extensively used to characterize the delta opioid receptor, they generally have low blood-brain permeability and poor metabolic stability. There are some reports that DPDPE and deltorphins can cross the blood-brain barrier (Williams et al., 1996; Fiori et al., 1997); and there has also been some effort to make more stable peptidomimetics (Rochon et al., 2013). In addition, considerable effort has been expended in developing small molecule (non-peptide) agonists to study the delta opioid receptor. Among these small molecule agonists SNC80, ARM390 and TAN67 are commonly used.
SNC80 was derived from the highly potent delta agonist (+)BW373U86 (Calderon et al., 1994), and is the most widely used systemically available small molecule delta agonist. In vitro, the Ki of SNC80 is in the low nM range, and it has similar selectivity to deltorphin II, as compared to mu (~200 fold) and kappa (~2500 fold) opioid receptors (Bilsky et al., 1995; Payza, 2004). SNC80 is usually administered subcutaneously or intraperitoneally, and is not highly effective when given per os (AstraZeneca personal communication). Unfortunately, SNC80 is difficult to dilute, and requires an acidic vehicle (Table 1). In our opinion, SNC80 acts as a bench-mark delta agonist, as it has a high selectivity for the delta opioid receptor and produces the widest array of receptor regulatory and signaling events, and behavioral effects.
ARM390 (Wei et al., 2000) is a highly selective delta opioid receptor agonist, with similar receptor binding and G protein activation properties to SNC80 (Wei et al., 2000; Marie et al., 2003a; Payza, 2004; Pradhan et al., 2009). It has a greater selectivity for the delta opioid receptor than SNC80, and affinity for mu and kappa ~4000 and ~6000 fold greater. Differences in the inter-nalization properties of SNC80 and ARM390 have been found; while SNC80 produces robust receptor intemalization, ARM390 does not induce substantial delta opioid receptor sequestration (Marie et al., 2003a; Pradhan et al., 2009); and this can have an impact on short- and long-term tolerance following chronic use (Pradhan et al., 2009, 2010). Although ARM390 is a low-internalizing agonist, it does appear to recruit arrestin 3; and this interaction is limited to the cell membrane (Pradhan et al., 2016). Importantly, ARM390 is only effective when administered per os by gavage (AstraZeneca, personal communication). Indeed, no analgesic effects to ARM390 were observed in the lactic acid-induced stretching assay in rats after its intraperitoneal administration (Negus et al., 2012).
TAN-67 is a morphinan derivative with high selectivity for the delta opioid receptor (Knapp et al., 1995). Although the racemic (+/−)-TAN-67 showed no analgesic activity, its enantiomer (−)-TAN-67 is highly effective (Nagase et al., 2001). TAN-67 has also been shown to be a low-internalizing delta agonist (Bradbury et al., 2009). Like most small molecule delta opioid receptor agonists, TAN-67 is systemically active and has been shown to be effective by icv, sc, it, and ip routes in different in vivo studies (Refs. in Table 1).
The extensive research carried out to discover non-peptide ligands for the delta opioid receptor has also provided highly selective antagonists. Naltrindole is the most commonly used delta opioid receptor antagonist (Portoghese et al., 1988). It has a high affinity for the delta opioid receptor, with a Ki of 0.18 nM for the human delta opioid receptor. However, naltrindole also has a low nanomolar affinity for the mu opioid receptor; and is only ~25-fold selective for delta over mu (Payza, 2004). Thus, at high concentration naltrindole will also bind to the mu opioid receptor. Other non-peptide antagonists are nal-triben (NTB), naltrindole 5′-isothyocianate (5′-NTII) and 7-benzylidenenaltrexone (BNTX), which have been used to study the putative delta opioid receptor subtypes (δ1 and δ2). Most of the pharmacological evidences for the existence of delta opioid receptor subtypes comes from pharmacological studies (Mattia et al., 1991; Sofuoglu et al., 1991; Maslov et al., 2009; Sugiyama et al., 2014; Beaudry et al., 2015). However, only one clone has been identified for the delta opioid receptor with no polymorphisms or viable splice variants (Gaveriaux-Ruff et al., 1997; Wei and Loh, 2002).
How to detect delta opioid receptors
Although there is a general consensus on the broad anatomical localization of delta opioid receptors (Mansour et al., 1988; Le Merrer et al., 2009), there has been much debate about the specific cell types that express these receptors (Bao et al., 2003; Guan et al., 2005; Cahill et al., 2007; Scherrer et al., 2009; Wang et al., 2010; Bardoni et al., 2014; Gendron et al., 2014). The main arguments are focused on which primary afferents delta opioid receptors are expressed on – small, peptidergic fibers that co-express mu opioid receptors (Bao et al., 2003; Zhang et al., 2006; Overland et al., 2009; Riedl et al., 2009; Wang et al., 2010; Chabot-Dore et al., 2015; Huang et al., 2015), versus medium/large fibers that are non-peptidergic with low co-expression with mu (Scherrer et al., 2009; Bardoni et al., 2014). This argument further extends to whether endogenous delta opioid receptors are located on intracellular vesicles (Svingos et al., 1998; Cahill et al., 2001, 2003, 2007; Wang and Pickel, 2001; Bao et al., 2003; Morinville et al., 2003, 2004; Gendron et al., 2006; Zhang et al., 2006; Kabli and Cahill, 2007; Wang et al., 2010; Huang et al., 2015) or are expressed on the cell membrane (Scherrer et al., 2009; Bardoni et al., 2014). The main discussion has centered on the best way to examine these receptors in fixed tissue. One issue that has plagued the field is the specificity of the antibodies used for traditional immunohistochemical detection of delta opioid receptors. Many of the commercially available antibodies that were cited in previous publications continued to show staining in delta opioid receptor knockout mice (Scherrer et al., 2009). Even in earlier studies examining the distribution of delta opioid receptor, a commonly used antibody (Dado et al., 1993; Arvidsson et al., 1995; Tao et al., 1998) was also found to be blocked by substance P (Arvidsson et al., 1995). Wang et al. have showed that two commercially available rabbit antibodies, used at very dilute concentrations, did not show staining in the exon 1 targeted delta opioid receptor knockout mouse (Wang et al., 2010). Confirmation of these results by other labs is still ongoing. To add additional complexity, there is batch variation in antibody production, so an antibody that was specific in one lot may no longer be in subsequent lots. Furthermore, variation in species, strain, and how the tissue is processed also contributes to the variability across labs. Antibodies to the delta opioid receptor may be used in immunoblotting protocol (Billa et al., 2010); and there are also antibodies targeting the serine 363 phosphory-lated form of the delta opioid receptor (Pradhan et al., 2009), which may also be adapted for immunostaining (Faget et al., 2012). In addition, subtractive immunization techniques have been used to generate antibodies specific to the mu-delta heterodimer (Gomes et al., 2014). Unfortunately, the lack of an unequivocal delta opioid receptor specific antibody has limited our ability to definitively characterize the physiology and expression of this receptor, especially in the translation of preclinical rodent data to human post-mortem tissue.
Another approach to visualizing the delta opioid receptor in fixed tissue has been to use a knockin mouse strategy in which the endogenous delta opioid receptor has been replaced with a fluorescent tagged receptor (DOR-eGFP), where eGFP is linked to the C-terminal tail ((Scherrer et al., 2006; Pradhan et al., 2015), see below and Table 2). These mice show fidelity to wildtype animals in terms of overall anatomical expression of delta opioid receptors as determined by in situ hybridization and radioligand binding (Bardoni et al., 2014). In addition, these mice have similar behavioral responses to C57BL/6 mice when treated with delta agonists (Pradhan et al., 2010). They are also an excellent tool to study the in vivo trafficking of the delta opioid receptor (Scherrer et al., 2006; Pradhan et al., 2009, 2010; Faget et al., 2012; Bertran-Gonzalez et al., 2013). However, there has been much debate about whether the addition of the eGFP tag alters the cellular distribution and subcellular compartmentalization of delta opioid receptors compared to the endogenous form (Gendron et al., 2014; Zhang et al., 2015). Controversy in this regard is due to the differences in the localization of delta opioid receptors found using DOR-eGFP mice versus delta opioid receptor antibodies. In dorsal root ganglia (DRG) and lumbar spinal cord, very few DOR-eGFP receptors were co-localized with mu opioid receptors (Scherrer et al., 2009), which is in contrast to studies performed with antibodies that indicate a physical and functional interaction between both receptors (see (Gendron et al., 2014) for a review). In addition, DOR-eGFP receptors are expressed on the cell membrane, in opposition to the findings that delta opioid receptors are on intracellular vesicles which are trafficked to the cell surface by release of substance P (Bao et al., 2003; Guan et al., 2005). Further investigation into these contentious issues is ongoing, and it is likely that under certain conditions different aspects of all of these findings are possible. For instance, multiple lines of evidence indicate that the delta opioid receptor is dynamic, and increased functionality is observed following chronic stimuli such as pain (Cahill et al., 2003; Kabli and Cahill, 2007; Pradhan et al., 2013), morphine (Cahill et al., 2001), and ethanol exposure (van Rijn et al., 2012). This receptor plasticity may also enhance mu-delta interactions. More importantly, a greater emphasis must be placed on the translational implications of these findings, especially considering that there are major differences in the expression of delta opioid receptor in the DRG and spinal cord of rodents versus primates and humans (Mennicken et al., 2003).
Table 2.
Mutant mouse models to study the delta opioid receptor
There are many alternative approaches to visualizing the delta opioid receptor, although these approaches do not generally allow for co-localization or cellular characterization. Autoradiography was used early on to determine the central and spinal regions that express delta opioid receptors (Mansour et al., 1988; Le Merrer et al., 2009). Commercially available radioligands include the agonists: [3H] or [125l]Deltorphin II, [3H]DADLE, and [3H]DPDPE; and the antagonist [3H]naltrindole. The radi-olabeled delta agonists may also bind to the mu opioid receptor (Fraser et al., 2000b; Payza, 2004; Scherrer et al., 2004), but can be overcome with the addition of the mu antagonist CTOP to the binding buffer (Pradhan and Clarke, 2005a).
Another approach which can minimize non-specific binding, and allows for electron microscopy is the use of photoaffinity radioligands. Photoaffinity labels can be covalently bound to the receptor by UV irradiation after washing away ligand that is free or weakly bound to non-specific targets. However, these ligands usually render a very low percentage of covalent incorporation (5–20%). The photoaffinity radioligands [3H]azido-DTLET and [25l]azido-DTLET have been used to study the autoradiographic localization of the delta opioid receptor in rat brain sections and membrane homogenates (Zajac et al., 1987; Bochet et al., 1988; Pasquini et al., 1992, 1993); and at the [25l]azido-DTLET has been used to detect delta opioid receptors at the ultrastructural level (Pasquini et al., 1992).
One of the more effective ways to examine delta opioid receptor expression and trafficking is the use of fluorescently labeled delta ligands. In comparison with radioligands, fluorescent probes are safer and allow receptor localization with enhanced temporal and spatial resolution (Handl et al., 2005; Emami-Nemini et al., 2013). However, fluorophores are bulky and can influence the binding properties of the ligand; moreover, they have hydrophobic properties that can affect their biological activity and selectivity (Kolb et al., 1983; Balboni et al., 2004). Nevertheless, a wide range of fluorescent conjugates of delta agonists and antagonists have been synthesized and are commercially available (Cell Aura Technologies Ltd. and Cisbio Bioassays). Fluorescent antagonists have proved useful to examine delta opioid receptor distribution in living cells (Kshirsagar et al., 1998; Balboni et al., 2004; Cohen et al., 2016); while agonists have been used to study delta opioid receptor inter-nalization and trafficking in vivo (Arttamangkul et al., 2000; Cahill et al., 2001; Lee et al., 2002).
Functional imaging techniques have also been used to study delta opioid receptor distribution, and specific radioligands for positron emission tomography (PET) are available. A potent and selective delta opioid receptor antagonist coupled to [11C]iodomethane, [11C]-N1′-methylnaltrindole, has been successfully used in several PET studies in rodents and humans (Lever et al., 1992; Madar et al., 2007; Weerts et al., 2008, 2011; Wand et al., 2013). The non-selective agonist [11C]diprenorphine that labels mu, kappa and delta opioid receptors has also been used to examine the distribution of opioid receptors in a variety of clinical conditions, including epilepsy (Hammers et al., 2007; McGinnity et al., 2013), pain (Jones et al., 2004; Willoch et al., 2004), and opioid addiction (Melichar et al., 2005; Williams et al., 2009). Taking into consideration the potential of PET to advance our understanding of the opioid system in the normal and diseased brain in vivo, considerable efforts are being made to increase the number of radiotracers designed to study the delta opioid receptor (Clayson et al., 2001; Tyacke et al., 2002).
THINGS TO CONSIDER WHEN EXAMINING THE BEHAVIORAL EFFECTS OF DELTA AGONISTS
Treatment of chronic pain, anxiety and depression are some of the primary behaviors in which delta agonists have been shown to be effective. However, they are also involved in a number of other behavioral effects including regulation of other drugs of abuse (Bosse et al., 2008; van Rijn et al., 2010; Pradhan et al., 2011a). Delta opioid agonists are not very effective in acute pain models (Gallantine and Meert, 2005), although they do work in the paw pressure assay (Fraser et al., 2000b). Delta agonists are effective in chemically-induced pain such as the formalin (for a review see (Gaveriaux-Ruff and Kieffer, 2011)), and acetic acid writhing tests (Gallantine and Meert, 2005). Animal models of chronic pain is where delta agonists have shown the most promise. They are highly effective in the Complete Freunds’ Adjuvent (CFA) model of inflammatory pain (Gendron et al., 2006; Pradhan et al., 2009), peripheral neuropathy models (Kabli and Cahill, 2007; Nozaki et al., 2014), and bone cancer-induced pain models (Otis et al., 2011). In addition, they have recently been shown to work in a model of migraine pain and aura (Pradhan et al., 2014; Charles and Pradhan, 2016).
The effectiveness of delta opioid receptor agonists in models of anxiety and depression has led to the development of these compounds for the treatment of emotional disorders. They have been highly effective in the elevated plus maze, forced swim test, tail suspension test, and social interaction tests (Broom et al., 2002a; Saitoh et al., 2005; Perrine et al., 2006). One caveat to consider when testing delta opioid receptor agonists is that some of them can cause locomotor stimulation, which can interfere with assays such as the elevated plus maze. For this reason, agonists such as SNC80, are often given subcutaneously in these paradigms and testing occurs at 1 h post-drug, after the loco-motor stimulant effect has diminished. There is very little expression of delta opioid receptors in brain stem regions that regulate nociception, and a high expression in regions that regulate emotional responses to pain (Pradhan and Clarke, 2005b; Pradhan et al., 2011b). Thus, the true potential for the therapeutic use of delta agonists may lie in the treatment of chronic pain conditions that have a significant co-morbidity with emotional dysregulation, such as fibromyalgia and migraine.
A further adverse effect of delta agonists is that some of them can also produce convulsions, and this has limited their development for clinical use. The pro-convulsant effect of delta agonists is ligand specific, and is a clear example of ligand-directed signaling or functional selectivity at the delta opioid receptor (Pradhan et al., 2012; Chu Sin Chung et al., 2014). SNC80 and (+)BW373U86 produce convulsions, however, most other systemically available agonists do not (Pradhan et al., 2011a). This deleterious effect is dependent on the activation of the delta opioid receptor (Comer et al., 1993; Broom et al., 2002b), and has been observed in mice, rats, and squirrel and rhesus monkeys (Dykstra et al., 1993; Broom et al., 2002a; Danielsson et al., 2006; Jutkiewicz et al., 2006). The convulsions produced by delta agonists like SNC80 or (+)BW373U86 are behaviorally similar, but weaker than those produced by 4 the epileptogenic drug pentylenetetrazol (Comer et al., 1993). Delta agonist-induced convulsions may be related to absence seizures in humans (Jutkiewicz et al., 2006). Convulsions produced by delta agonists generally occur within 2–5 min of subcutaneous injection, and are characterized by clonic movements of the head, face and fore-paws and potentially extension of the body (Comer et al., 1993). The convulsant period is of very short duration, and can be completed in less than 5–10 min post-injection. Therefore, it is imperative to pay close attention to animals following administration of delta agonist as these convulsions are easy to miss. Convulsions are followed by catalepsy, which is a brief period lasting 10–30 min post-injection of delta agonist, where the animal shows muscular rigidity and lack of reaction to external stimuli. Catalepsy can be determined using the bar test, which consists on placing the forepaws of the animals on a bar suspended approximately 1 inch above the ground. Animals are considered cataleptic when they fail to remove their forepaws within 15 s (Comer et al., 1993). Following delta agonist-induced convulsion and catalepsy, rodents circumambulate normally and are indistinguishable from naive controls (Comer et al., 1993; Jutkiewicz et al., 2006).
Genetic tools to study the delta opioid receptor
Over the past 30 years, genetic mouse models have provided a novel platform in which to explore the role of delta opioid receptor in vivo. Here we discuss genetic mouse models that serve as suitable tools to further investigate the delta opioid receptor (Table 2).
Delta opioid receptor-KOexon 2
There are four different delta opioid receptor knockout animals (Zhu et al., 1999; Filliol et al., 2000). Although they show similar phenotypes, there are some important differences between them. In general, delta opioid receptor knockout mice show no apparent developmental deficits, are similar in size to their wild-type littermates, and both sexes are reproductively fertile. John Pintar’s group developed a knockout of the delta opioid receptor by deleting exon 2 of the Oprd1 gene in 1999 (Zhu et al., 1999) (Table 2). These mice showed no binding to both [3H]DPDPE or [3H]Deltorphin II, and neither agonist produced pain-relieving effects when injected intrathecally. However, intracerebroventricular injection of DPDPE and deltrophin II to these knockouts continued to produce an antinociceptive effect, which may be due to activation of non-delta opioid receptors (Zhu et al., 1999). In 2009, Jennifer Whistler’s group also produced an exon 2 knockout of the delta opioid receptor using a bacterial artificial chromosome (BAC) transgenic approach (van Rijn and Whistler, 2009a). In the process, this group also obtained “floxed” DOR mice, in which exon 2 is flanked by 2 loxP sites; which can be used to generate conditional knockouts ((van Rijn and Whistler, 2009a), and see below).
Delta opioid receptor-KOexon 1
Another delta opioid receptor knockout mouse was generated by Brigitte Kieffer’s lab, and in this case exon 1 of the Oprdl gene was targeted (Filliol et al., 2000) (Table 2). Similar to the delta opioid receptor-KOexon 2 mice, delta opioid receptor-KOexon 1 showed no binding to [3H]Naltrindole, [3H]DPDPE, and [3H]Deltorphin in the brain or periphery. Interestingly, quantitative receptor autoradiography shows compensatory down-regulation of mu and changes in kappa opioid receptor expression in the homozygous mutant mice, but not heterozygous controls (Goody et al., 2002). These mice were used to demonstrate that delta opioid receptors regulate emotional tone, and they show higher responses in tests of anxiety and depression as well as increase locomotor activity and impaired learning (Filliol et al., 2000; Kieffer and Gaveriaux-Ruff, 2002; Le Merrer et al., 2011). In addition, these mice show enhanced sensitivity in models of chronic pain (Gaveriaux-Ruff et al., 2011). For a summary of the changes that occur in delta opioid receptor knockout animals we refer the reader to the following reviews (Kieffer and Gaveriaux-Ruff, 2002; Le Merrer et al., 2009).
DOR-eGFP knockin mice
DOR-eGFP mice are knockin mice in which the endogenous delta opioid receptor is replaced by delta opioid receptor fused at the C terminus with enhanced green fluorescent protein (eGFP) (Scherrer et al., 2006) (Table 2). Although delta opioid receptor transcription remains functional in DOR-eGFP mice, quantitative mRNA analyses show a ~50% increase of Oprd1 transcription when compared to wild-type littermates (Scherrer et al., 2006). DOR-eGFP fluorescence can be detected as early as postnatal day 3, reaching maximal intensity throughout the entire brain, spinal cord, and dorsal root ganglia by postnatal day 15 (Scherrer et al., 2006). These mice have been used to determine receptor expression (Scherrer et al., 2009; Poole et al., 2011; Erbs et al., 2012; Bardoni et al., 2014), and in vivo receptor trafficking to both exogenous (Pradhan et al., 2009, 2010) and endogenous (Poole et al., 2011; Faget et al., 2012; Bertran-Gonzalez et al., 2013) stimuli. These mice have also been crossed with the fluorescent mu-Cherry knockin mice to examine co-expression of these two receptors (Erbs et al., 2015). Several considerations are associated with these mice, as discussed in the detection section.
CONDITIONAL KNOCKOUT MOUSE MODELS
DORCMV cKO
Conditional knockouts (cKOs) of the delta opioid receptor have been generated using the Cre-loxP system. Floxed delta opioid receptor (Oprd1fl/fl) mice were generated by introducing two loxP sites into exon 2 (Gaveriaux-Ruff et al., 2011). In order to assess the functionality of the loxP sites in Oprd1fl/fl mice they were crossed with a constitutively expressing Cre line ((cytomegalovirus) CMV-Cre) (Gaveriaux-Ruff et al., 2011). These DORCMV cKO mice are thus a fourth delta opioid receptor knockout mouse, in which there is a complete deletion of delta opioid receptor from brain and periphery, as well as an enhanced anxiety phenotype (Gaveriaux-Ruff et al., 2011; Chu Sin Chung et al., 2015).
DORNav1.8 cKO
Oprd1fl/fl mice were crossed with a Nav1.8-Cre line to generate a peripherally restricted knockout of delta opioid receptors (Gaveriaux-Ruff et al., 2011). DORNav1 8 cKO show a ~60–70% decrease in delta opioid receptors in the small and medium dorsal root ganglia, but otherwise brain and spinal delta opioid receptor expression remains intact (Table 2). These mice show no change in responses to acute noxious heat and mechanical stimuli. However in a model of chronic inflammatory pain; mechanical, but not heat, hyperalgesia is altered in DORNav1.8 cKO interestingly, the anti-hyperalgesic effects of SNC80 are lost in inflammatory and neuropathic pain conditions, but this agonist continues to inhibit formalin-induced pain (Gaveriaux-Ruff et al., 2011). One caution with these cKO mice, is that the Nav1.8 Cre line may not be restricted to nociceptors, and may also delete delta opioid receptors in ganglia involved with mechanosensation (Shields et al., 2012).
DORDlx5/6 cKQ
Floxed delta opioid receptor mice have also been bred with the Dlx5/6-Cre line (Chu Sin Chung et al., 2014, 2015), a line in which Cre is expressed in forebrain GABAergic neurons (Ruest et al., 2003; Monory et al., 2006). DORDlx5/6 cKO show a near total loss of delta opioid receptors in the olfactory bulb, nucleus accumbens and caudate putamen, as well as a ~50% loss in the hippocampus. Delta opioid receptor levels remain unchanged in the midbrain, brain stem, and spinal cord (Chu Sin Chung et al., 2014, 2015) (Table 2). Interestingly, DORDlx5/6 cKO display reduced levels of anxiety and depression in contrast to the constitutive delta opioid receptor knockout (Chu Sin Chung et al., 2015). These mice have also been used to show that SNC80 produces convulsions through disinhibition of forebrain GABAergic neurons (Chu Sin Chung et al., 2014). One caveat when generating these mice is to breed male Dlx5/6-Cre mice with female floxed mice, as breeding with the female Cre will result in constitutive knockout.
Future work with currently available mutant mice, and novel genetic tools will continue to shed light on the role of delta opioid receptors in diverse biological conditions.
CONCLUSIONS
The delta opioid receptor, since it’s cloning in the early 1990s, has been pursued as a therapeutic target for the treatment of chronic pain. Unfortunately, clinical trials of delta agonists for this indication have not yielded positive outcomes (https://clinicaltrials.gov/ct2/show/results/NCT00979953). Although a number of factors can account for this lack of success, these disappointing results have dampened enthusiasm for this receptor in the treatment of pain disorders. The phenotype of delta opioid receptor knockouts (Filliol et al., 2000), and subsequent discovery that delta agonists have anxiolytic and anti-depressant effects (Broom et al., 2002a; Saitoh et al., 2005; Perrine et al., 2006) moved the development of delta agonists toward the treatment of emotional disorders. Recent phase I and IIa clinical trial tested the delta agonists AZD2327 (Richards et al., 2016) and AZD7268 (https://clinicaltrials.gov/ct2/results?term=AZD7268) in anxious major depressive disorder. Although neither study reached primary endpoints for depression or anxiety, in both cases, analysis of secondary endpoints indicated potential benefit for anxiety. In addition, recent preclinical work indicates that delta agonists may be effective for the treatment of migraine (Pradhan et al., 2014), which opens yet another avenue to the therapeutic benefit of this receptor. Considering that the delta opioid receptor is less studied than the mu opioid receptor, there are an incredible number of well-validated tools available to probe delta opioid receptor function. The ongoing development of additional tools, and encouragement of scientists from other fields to examine the delta opioid receptor in their models, will enhance the clinical potential of this receptor.
Highlights.
Tools available to study the delta opioid receptor are reviewed.
Cell-based systems to study delta opioid receptor are discussed.
Behavioral effects and limitations of delta agonists are reviewed.
Mutant mouse models to study in vivo delta opioid receptor action are described.
Acknowledgments
This work was supported by NIH-NIDA grant DA031243, the Department of Defense PR141746, and the Department of Psychiatry UIC.
REFERENCES
- Allouche S, Polastron J, Hasbi A, Homburger V, Jauzac P. Differential G-protein activation by alkaloid and peptide opioid agonists in the human neuroblastoma cell line SK-N-BE. Biochem J. 1999a;342(Pt 1):71–78. [PMC free article] [PubMed] [Google Scholar]
- Allouche S, Roussel M, Marie N, Jauzac P. Differential desensitization of human delta-opioid receptors by peptide and alkaloid agonists. Eur J Pharmacol. 1999b;371:235–240. doi: 10.1016/s0014-2999(99)00180-6. [DOI] [PubMed] [Google Scholar]
- Allouche S, Hasbi A, Ferey V, Sola B, Jauzac P, Polastron J. Pharmacological delta1- and delta2-opioid receptor subtypes in the human neuroblastoma cell line SK-N-BE: no evidence for distinct molecular entities. Biochem Pharmacol. 2000;59:915–925. doi: 10.1016/s0006-2952(99)00404-9. [DOI] [PubMed] [Google Scholar]
- Alt A, Clark MJ, Woods JH, Traynor JR. Mu and Delta opioid receptors activate the same G proteins in human neuroblastoma SH-SY5Y cells. Br J Pharmacol. 2002;135:217–225. doi: 10.1038/sj.bjp.0704430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arttamangkul S, Alvarez-Maubecin V, Thomas G, Williams JT, Grandy DK. Binding and internalization of fluorescent opioid peptide conjugates in living cells. Mol Pharmacol. 2000;58:1570–1580. doi: 10.1124/mol.58.6.1570. [DOI] [PubMed] [Google Scholar]
- Arvidsson U, Dado RJ, Riedl M, Lee JH, Law PY, Loh HH, Elde R, Wessendorf MW. Delta-Opioid receptor immunoreactivity: distribution in brainstem and spinal cord, and relationship to biogenic amines and enkephalin. J Neurosci. 1995;15:1215–1235. doi: 10.1523/JNEUROSCI.15-02-01215.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atwood BK, Lopez J, Wager-Miller J, Mackie K, Straiker A. Expression of G protein-coupled receptors and related proteins in HEK293, AtT20, BV2, and N18 cell lines as revealed by microarray analysis. BMC Genom. 2011;12:14. doi: 10.1186/1471-2164-12-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Audet N, Gales C, Archer-Lahlou E, Vallieres M, Schiller PW, Bouvier M, Pineyro G. Bioluminescence resonance energy transfer assays reveal ligand-specific conformational changes within preformed signaling complexes containing delta-opioid receptors and heterotrimeric G proteins. J Biol Chem. 2008;283:15078–15088. doi: 10.1074/jbc.M707941200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Audet N, Charfi I, Mnie-Filali O, Amraei M, Chabot-Dore AJ, Millecamps M, Stone LS, Pineyro G. Differential association of receptor-Gbetagamma complexes with beta-arrestin2 determines recycling bias and potential for tolerance of delta opioid receptor agonists. J Neurosci. 2012;32:4827–4840. doi: 10.1523/JNEUROSCI.3734-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balboni G, Salvadori S, Dal Piaz A, Bortolotti F, Argazzi R, Negri L, Lattanzi R, Bryant SD, Jinsmaa Y, Lazarus LH. Highly selective fluorescent analogue of the potent delta-opioid receptor antagonist Dmt-Tic. J Med Chem. 2004;47:6541–6546. doi: 10.1021/jm040128h. [DOI] [PubMed] [Google Scholar]
- Bao L, Jin SX, Zhang C, Wang LH, Xu ZZ, Zhang FX, Wang LC, Ning FS, Cai HJ, Guan JS, Xiao HS, Xu ZQ, He C, Hokfelt T, Zhou Z, Zhang X. Activation of delta opioid receptors induces receptor insertion and neuropeptide secretion. Neuron. 2003;37:121–133. doi: 10.1016/s0896-6273(02)01103-0. [DOI] [PubMed] [Google Scholar]
- Bardoni R, Tawfik VL, Wang D, Francois A, Solorzano C, Shuster SA, Choudhury P, Betelli C, Cassidy C, Smith K, de Nooij JC, Mennicken F, O’Donnell D, Kieffer BL, Woodbury CJ, Basbaum Al, MacDermott AB, Scherrer G. Delta opioid receptors presynaptically regulate cutaneous mechanosensory neuron input to the spinal cord dorsal horn. Neuron. 2014;81:1312–1327. doi: 10.1016/j.neuron.2014.01.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baumhaker Y, Gafni M, Keren O, Same Y. Selective and interactive down-regulation of mu- and delta-opioid receptors in human neuroblastoma SK-N-SH cells. Mol Pharmacol. 1993;44:461–467. [PubMed] [Google Scholar]
- Beaudry H, Gendron L, Moron JA. Implication of delta opioid receptor subtype 2 but not delta opioid receptor subtype 1 in the development of morphine analgesic tolerance in a rat model of chronic inflammatory pain. Eur J Neurosci. 2015;41:901–907. doi: 10.1111/ejn.12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beddell CR, Clark RB, Lowe LA, Wilkinson S, Chang KJ, Cuatrecasas P, Miller R. A conformational analysis for leucine-enkephalin using activity and binding data of synthetic analogues. Br J Pharmacol. 1977;61:351–356. doi: 10.1111/j.1476-5381.1977.tb08427.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertran-Gonzalez J, Laurent V, Chieng BC, Christie MJ, Balleine BW. Learning-related translocation of delta-opioid receptors on ventral striatal cholinergic interneurons mediates choice between goal-directed actions. J Neurosci. 2013;33:16060–16071. doi: 10.1523/JNEUROSCI.1927-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Billa SK, Xia Y, Moron JA. Disruption of morphine-conditioned place preference by a delta2-opioid receptor antagonist: study of mu-opioid and delta-opioid receptor expression at the synapse. Eur J Neurosci. 2010;32:625–631. doi: 10.1111/j.1460-9568.2010.07314.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bilsky EJ, Calderon SN, Wang T, Bernstein RN, Davis P, Hruby VJ, McNutt RW, Rothman RB, Rice KC, Porreca F. SNC 80, a selective, nonpeptidic and systemically active opioid delta agonist. J Pharmacol Exp Ther. 1995;273:359–366. [PubMed] [Google Scholar]
- Bilsky EJ, Qian X, Hruby VJ, Porreca F. Antinociceptive activity of [beta-methyl-2′,6′-dimethyltyrosine(1)]-substituted cyclic [d-Pen(2), d-Pen(5)]Enkephalin and [d-Ala(2), Asp(4)] Deltorphin analogs. J Pharmacol Exp Ther. 2000;293:151–158. [PubMed] [Google Scholar]
- Bochet P, Icard-Liepkalns C, Pasquini F, Garbay-Jaureguiberry C, Beaudet A, Roques B, Rossier J. Photoaffinity labeling of opioid delta receptors with an iodinated azido-ligand: [125I][d-Thr2, pN3Phe4, Leu5]enkephalyl-Thr6. Mol Pharmacol. 1988;34:436–443. [PubMed] [Google Scholar]
- Bosse KE, Jutkiewicz EM, Gnegy ME, Traynor JR. The selective delta opioid agonist SNC80 enhances amphetamine-mediated efflux of dopamine from rat striatum. Neuropharmacology. 2008;55:755–762. doi: 10.1016/j.neuropharm.2008.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bradbury FA, Zelnik JC, Traynor JR. G protein independent phosphorylation and internalization of the delta-opioid receptor. J Neurochem. 2009;109:1526–1535. doi: 10.1111/j.1471-4159.2009.06082.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brandt MR, Furness MS, Rice KC, Fischer BD, Negus SS. Studies of tolerance and dependence with the delta-opioid agonist SNC80 in rhesus monkeys responding under a schedule of food presentation. J Pharmacol Exp Ther. 2001;299:629–637. [PubMed] [Google Scholar]
- Broom DC, Jutkiewicz EM, Folk JE, Traynor JR, Rice KC, Woods JH. Nonpeptidic delta-opioid receptor agonists reduce immobility in the forced swim assay in rats. Neuropsychopharmacology. 2002a;26:744–755. doi: 10.1016/S0893-133X(01)00413-4. [DOI] [PubMed] [Google Scholar]
- Broom DC, Nitsche JF, Pintar JE, Rice KC, Woods JH, Traynor JR. Comparison of receptor mechanisms and efficacy requirements for delta-agonist-induced convulsive activity and antinociception in mice. J Pharmacol Exp Ther. 2002b;303:723–729. doi: 10.1124/jpet.102.036525. [DOI] [PubMed] [Google Scholar]
- Cahill CM, Morinville A, Lee MC, Vincent JP, Collier B, Beaudet A. Prolonged morphine treatment targets delta opioid receptors to neuronal plasma membranes and enhances delta-mediated antinociception. J Neurosci. 2001;21:7598–7607. doi: 10.1523/JNEUROSCI.21-19-07598.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cahill CM, Morinville A, Hoffert C, O’Donnell D, Beaudet A. Up-regulation and trafficking of delta opioid receptor in a model of chronic inflammation: implications for pain control. Pain. 2003;101:199–208. doi: 10.1016/s0304-3959(02)00333-0. [DOI] [PubMed] [Google Scholar]
- Cahill CM, Holdridge SV, Morinville A. Trafficking of delta-opioid receptors and other G-protein-coupled receptors: implications for pain and analgesia. Trends Pharmacol Sci. 2007;28:23–31. doi: 10.1016/j.tips.2006.11.003. [DOI] [PubMed] [Google Scholar]
- Calderon SN, Rothman RB, Porreca F, Flippen-Anderson JL, McNutt RW, Xu H, Smith LE, Bilsky EJ, Davis P, Rice KC. Probes for narcotic receptor mediated phenomena. 19. Synthesis of (+)-4-[(alpha R)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N, N-diethylbenzamide (SNC 80): a highly selective, nonpeptide delta opioid receptor agonist. J Med Chem. 1994;37:2125–2128. doi: 10.1021/jm00040a002. [DOI] [PubMed] [Google Scholar]
- Chabot-Dore AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and alpha2-adrenoceptors. Br J Pharmacol. 2015;172:388–402. doi: 10.1111/bph.12695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan AS, Law PY, Loh HH, Ho PN, Wu WM, Chan JS, Wong YH. The first and third intracellular loops together with the carboxy terminal tail of the delta-opioid receptor contribute toward functional interaction with Galpha16. J Neurochem. 2003;87:697–708. doi: 10.1046/j.1471-4159.2003.02040.x. [DOI] [PubMed] [Google Scholar]
- Chang KJ, Cuatrecasas P. Multiple opiate receptors. Enkephalins and morphine bind to receptors of different specificity. J Biol Chem. 1979;254:2610–2618. [PubMed] [Google Scholar]
- Charfi I, Nagi K, Mnie-Filali O, Thibault D, Balboni G, Schiller PW, Trudeau LE, Pineyro G. Ligand- and cell-dependent determinants of internalization and cAMP modulation by delta opioid receptor (DOR) agonists. Cell Mol Life Sci. 2014;71:1529–1546. doi: 10.1007/s00018-013-1461-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charfi I, Audet N, Bagheri Tudashki H, Pineyro G. Identifying ligand-specific signalling within biased responses: focus on delta opioid receptor ligands. Br J Pharmacol. 2015;172:435–448. doi: 10.1111/bph.12705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charles A, Pradhan AA. Delta-opioid receptors as targets for migraine therapy. Curr Opin Neurol. 2016;29:314–319. doi: 10.1097/WCO.0000000000000311. [DOI] [PubMed] [Google Scholar]
- Cheng J, Standifer KM, Tublin PR, Su W, Pasternak GW. Demonstration of kappa 3-opioid receptors in the SH-SY5Y human neuroblastoma cell line. J Neurochem. 1995;65:170–175. doi: 10.1046/j.1471-4159.1995.65010170.x. [DOI] [PubMed] [Google Scholar]
- Cheng ZJ, Fan GH, Zhao J, Zhang Z, Wu YL, Jiang LZ, Zhu Y, Pei G, Ma L. Endogenous opioid receptor-like receptor in human neuroblastoma SK-N-SH cells: activation of inhibitory G protein and homologous desensitization. NeuroReport. 1997;8:1913–1918. doi: 10.1097/00001756-199705260-00024. [DOI] [PubMed] [Google Scholar]
- Chiang T, Sansuk K, van Rijn RM. Beta-arrestin 2 dependence of delta opioid receptor agonists is correlated with alcohol intake. Br J Pharmacol. 2015 doi: 10.1111/bph.13374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu Sin Chung P, Kieffer BL. Delta opioid receptors in brain function and diseases. Pharmacol Ther. 2013;140:112–120. doi: 10.1016/j.pharmthera.2013.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu Sin Chung P, Boehrer A, Stephan A, Matifas A, Scherrer G, Darcq E, Befort K, Kieffer BL. Delta opioid receptors expressed in forebrain GABAergic neurons are responsible for SNC80-induced seizures. Behav Brain Res. 2014;278C:429–434. doi: 10.1016/j.bbr.2014.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu Sin Chung P, Keyworth HL, Martin-Garcia E, Charbogne P, Darcq E, Bailey A, Filliol D, Matifas A, Scherrer G, Ouagazzal AM, Gaveriaux-Ruff C, Befort K, Maldonado R, Kitchen I, Kieffer BL. A novel anxiogenic role for the delta opioid receptor expressed in GABAergic forebrain neurons. Biol Psychiatry. 2015;77:404–415. doi: 10.1016/j.biopsych.2014.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clayson J, Jales A, Tyacke RJ, Hudson AL, Nutt DJ, Lewis JW, Husbands SM. Selective delta-opioid receptor ligands: potential PET ligands based on naltrindole. Bioorg Med Chem Lett. 2001;11:939–943. doi: 10.1016/s0960-894x(01)00112-3. [DOI] [PubMed] [Google Scholar]
- Codd EE, Carson JR, Colburn RW, Stone DJ, Van Besien CR, Zhang SP, Wade PR, Gallantine EL, Meert TF, Molino L, Pullan S, Razler CM, Dax SL, Flores CM. JNJ-20788560 [9-(8-azabicyclo[3.2.1]oct-3-ylidene)-9H-xanthene-3-carboxylic acid diethylamide], a selective delta opioid receptor agonist, is a potent and efficacious antihyperalgesic agent that does not produce respiratory depression, pharmacologic tolerance, or physical dependence. J Pharmacol Exp Ther. 2009;329:241–251. doi: 10.1124/jpet.108.146969. [DOI] [PubMed] [Google Scholar]
- Cohen AS, Patek R, Enkemann SA, Johnson JO, Chen T, Toloza E, Vagner J, Morse DL. Delta-opioid receptor (deltaOR) targeted near-infrared fluorescent agent for imaging of lung cancer: synthesis and evaluation in vitro and in vivo. Bioconjug Chem. 2016;27:427–438. doi: 10.1021/acs.bioconjchem.5b00516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Comer SD, Hoenicke EM, Sable Al, McNutt RW, Chang KJ, De Costa BR, Mosberg HI, Woods JH. Convulsive effects of systemic, administration of the delta opioid agonist BW373U86 in mice. J Pharmacol Exp Ther. 1993;267:888–895. [PubMed] [Google Scholar]
- Cone Rl, Lameh J, Sadee W. Rapid agonist-induced loss of 125I–beta-endorphin opioid receptor sites in NG108-15, but not SK-N-SH neuroblastoma cells. Life Sci. 1991;49:PL147–PL152. doi: 10.1016/0024-3205(91)90396-s. [DOI] [PubMed] [Google Scholar]
- Dado RJ, Law PY, Loh HH, Elde R. Immunofluorescent identification of a delta (delta)-opioid receptor on primary afferent nerve terminals. NeuroReport. 1993;5:341–344. doi: 10.1097/00001756-199312000-00041. [DOI] [PubMed] [Google Scholar]
- Danielsson I, Gasior M, Stevenson GW, Folk JE, Rice KC, Negus SS. Electroencephalographic and convulsant effects of the delta opioid agonist SNC80 in rhesus monkeys. Pharmacol Biochem Behav. 2006;85:428–434. doi: 10.1016/j.pbb.2006.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dykstra LA, Schoenbaum GM, Yarbrough J, McNutt R, Chang KJ. A novel delta opioid agonist, BW373U86, in squirrel monkeys responding under a schedule of shock titration. J Pharmacol Exp Ther. 1993;267:875–882. [PubMed] [Google Scholar]
- Elliott J, Guo L, Traynor JR. Tolerance to mu-opioid agonists in human neuroblastoma SH-SY5Y cells as determined by changes in guanosine-5′-O-(3-[35S]-thio)triphosphate binding. Br J Pharmacol. 1997;121:1422–1428. doi: 10.1038/sj.bjp.0701253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emami-Nemini A, Roux T, Leblay M, Bourrier E, Lamarque L, Trinquet E, Lohse MJ. Time-resolved fluorescence ligand binding for G protein-coupled receptors. Nat Protoc. 2013;8:1307–1320. doi: 10.1038/nprot.2013.073. [DOI] [PubMed] [Google Scholar]
- Erbs E, Faget L, Scherrer G, Kessler P, Hentsch D, Vonesch JL, Matifas A, Kieffer BL, Massotte D. Distribution of delta opioid receptor-expressing neurons in the mouse hippocampus. Neuroscience. 2012;221:203–213. doi: 10.1016/j.neuroscience.2012.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erbs E, Faget L, Scherrer G, Matifas A, Filliol D, Vonesch JL, Koch M, Kessler P, Hentsch D, Birling MC, Koutsourakis M, Vasseur L, Veinante P, Kieffer BL, Massotte D. A mu-delta opioid receptor brain atlas reveals neuronal co-occurrence in subcortical networks. Brain Struct Funct. 2015;220:677–702. doi: 10.1007/s00429-014-0717-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erspamer V, Melchiorri P, Falconieri-Erspamer G, Negri L, Corsi R, Severini C, Barra D, Simmaco M, Kreil G. Deltorphins: a family of naturally occurring peptides with high affinity and selectivity for delta opioid binding sites. Proc Natl Acad Sci U S A. 1989;86:5188–5192. doi: 10.1073/pnas.86.13.5188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans CJ, Keith DE, Jr, Morrison H, Magendzo K, Edwards RH. Cloning of a delta opioid receptor by functional expression. Science. 1992;258:1952–1955. doi: 10.1126/science.1335167. [DOI] [PubMed] [Google Scholar]
- Faget L, Erbs E, Le Merrer J, Scherrer G, Matifas A, Benturquia N, Noble F, Decossas M, Koch M, Kessler P, Vonesch JL, Schwab Y, Kieffer BL, Massotte D. In vivo visualization of delta opioid receptors upon physiological activation uncovers a distinct internalization profile. J Neurosci. 2012;32:7301–7310. doi: 10.1523/JNEUROSCI.0185-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fan SF, Shen KF, Scheideler MA, Crain SM. F11 neuroblastoma x DRG neuron hybrid cells express inhibitory mu- and delta-opioid receptors which increase voltage-dependent K+ currents upon activation. Brain Res. 1992;590:329–333. doi: 10.1016/0006-8993(92)91116-v. [DOI] [PubMed] [Google Scholar]
- Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F, Befort K, Gaveriaux-Ruff C, Dierich A, LeMeur M, Valverde O, Maldonado R, Kieffer BL. Mice deficient for delta- and mu-opioid receptors exhibit opposing alterations of emotional responses. Nat Genet. 2000;25:195–200. doi: 10.1038/76061. [DOI] [PubMed] [Google Scholar]
- Fiori A, Cardelli P, Negri L, Savi MR, Strom R, Erspamer V. Deltorphin transport across the blood-brain barrier. Proc Natl Acad Sci U S A. 1997;94:9469–9474. doi: 10.1073/pnas.94.17.9469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francel PC, Harris K, Smith M, Fishman MC, Dawson G, Miller RJ. Neurochemical characteristics of a novel dorsal root ganglion X neuroblastoma hybrid cell line, F-11. J Neurochem. 1987;48:1624–1631. doi: 10.1111/j.1471-4159.1987.tb05711.x. [DOI] [PubMed] [Google Scholar]
- Fraser GL, Gaudreau GA, Clarke PB, Menard DP, Perkins MN. Antihyperalgesic effects of delta opioid agonists in a rat model of chronic inflammation. Br J Pharmacol. 2000a;129:1668–1672. doi: 10.1038/sj.bjp.0703248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fraser GL, Pradhan AA, Clarke PB, Wahlestedt C. Supraspinal antinociceptive response to [d-Pen(2,5)]-enkephalin (DPDPE) is pharmacologically distinct from that to other delta-agonists in the rat. J Pharmacol Exp Ther. 2000b;295:1135–1141. [PubMed] [Google Scholar]
- Fundytus ME, Schiller PW, Shapiro M, Weltrowska G, Coderre TJ. Attenuation of morphine tolerance and dependence with the highly selective delta-opioid receptor antagonist TIPP[psi] Eur J Pharmacol. 1995;286:105–108. doi: 10.1016/0014-2999(95)00554-x. [DOI] [PubMed] [Google Scholar]
- Gallantine EL, Meert TF. A comparison of the antinociceptive and adverse effects of the mu-opioid agonist morphine and the delta-opioid agonist SNC80. Basic Clin Pharmacol Toxicol. 2005;97:39–51. doi: 10.1111/j.1742-7843.2005.pto_97107.x. [DOI] [PubMed] [Google Scholar]
- Gaveriaux-Ruff C, Kieffer BL. Delta opioid receptor analgesia: recent contributions from pharmacology and molecular approaches. Behav Pharmacol. 2011;22:405–414. doi: 10.1097/FBP.0b013e32834a1f2c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaveriaux-Ruff C, Karchewski LA, Hever X, Matifas A, Kieffer BL. Inflammatory pain is enhanced in delta opioid receptor-knockout mice. Eur J Neurosci. 2008;27:2558–2567. doi: 10.1111/j.1460-9568.2008.06223.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaveriaux-Ruff C, Nozaki C, Nadal X, Hever XC, Weibel R, Matifas A, Reiss D, Filliol D, Nassar MA, Wood JN, Maldonado R, Kieffer BL. Genetic ablation of delta opioid receptors in nociceptive sensory neurons increases chronic pain and abolishes opioid analgesia. Pain. 2011 doi: 10.1016/j.pain.2010.12.031. [DOI] [PubMed] [Google Scholar]
- Gendron L, Lucido AL, Mennicken F, O’Donnell D, Vincent JP, Stroh T, Beaudet A. Morphine and pain-related stimuli enhance cell surface availability of somatic delta-opioid receptors in rat dorsal root ganglia. J Neurosci. 2006;26:953–962. doi: 10.1523/JNEUROSCI.3598-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gendron L, Mittal N, Beaudry H, Walwyn W. Recent advances on the delta opioid receptor: From trafficking to function. Br J Pharmacol. 2014 doi: 10.1111/bph.12706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geraghty RJ, Capes-Davis A, Davis JM, Downward J, Freshney Rl, Knezevic I, Lovell-Badge R, Masters JR, Meredith J, Stacey GN, Thraves P, Vias M, Cancer Research UK. Guidelines for the use of cell lines in biomedical research. Br J Cancer. 2014;111:1021–1046. doi: 10.1038/bjc.2014.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldstein A, Naidu A. Multiple opioid receptors: ligand selectivity profiles and binding site signatures. Mol Pharmacol. 1989;36:265–272. [PubMed] [Google Scholar]
- Gomes I, Jordan BA, Gupta A, Trapaidze N, Nagy V, Devi LA. Heterodimerization of mu and delta opioid receptors: a role in opiate synergy. J Neurosci. 2000;20:RC110. doi: 10.1523/JNEUROSCI.20-22-j0007.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomes I, Gupta A, Filipovska J, Szeto HH, Pintar JE, Devi LA. A role for heterodimerization of mu and delta opiate receptors in enhancing morphine analgesia. Proc Natl Acad Sci U S A. 2004;101:5135–5139. doi: 10.1073/pnas.0307601101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomes I, Gupta A, Bushlin I, Devi LA. Antibodies to probe endogenous G protein-coupled receptor heteromer expression, regulation, and function. Front Pharmacol. 2014;5:268. doi: 10.3389/fphar.2014.00268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goody RJ, Oakley SM, Filliol D, Kieffer BL, Kitchen I. Quantitative autoradiographic mapping of opioid receptors in the brain of delta-opioid receptor gene knockout mice. Brain Res. 2002;945:9–19. doi: 10.1016/s0006-8993(02)02452-6. [DOI] [PubMed] [Google Scholar]
- Guan JS, Xu ZZ, Gao H, He SQ, Ma GQ, Sun T, Wang LH, Zhang ZN, Lena I, Kitchen I, Elde R, Zimmer A, He C, Pei G, Bao L, Zhang X. Interaction with vesicle luminal protachykinin regulates surface expression of delta-opioid receptors and opioid analgesia. Cell. 2005;122:619–631. doi: 10.1016/j.cell.2005.06.010. [DOI] [PubMed] [Google Scholar]
- Hammers A, Asselin MC, Hinz R, Kitchen I, Brooks DJ, Duncan JS, Koepp MJ. Upregulation of opioid receptor binding following spontaneous epileptic seizures. Brain. 2007;130:1009–1016. doi: 10.1093/brain/awm012. [DOI] [PubMed] [Google Scholar]
- Hamprecht B, Glaser T, Reiser G, Bayer E, Propst F. Culture and characteristics of hormone-responsive neuroblastoma X glioma hybrid cells. Methods Enzymol. 1985;109:316–341. doi: 10.1016/0076-6879(85)09096-6. [DOI] [PubMed] [Google Scholar]
- Handl HL, Vagner J, Yamamura HI, Hruby VJ, Gillies RJ. Development of a lanthanide-based assay for detection of receptor-ligand interactions at the delta-opioid receptor. Anal Biochem. 2005;343:299–307. doi: 10.1016/j.ab.2005.05.040. [DOI] [PubMed] [Google Scholar]
- Henry AG, White IJ, Marsh M, von Zastrow M, Hislop JN. The role of ubiquitination in lysosomal trafficking of delta-opioid receptors. Traffic. 2011;12:170–184. doi: 10.1111/j.1600-0854.2010.01145.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heumann R, Valet G, Maison D, Kemper J, Reiser G, Hamprecht B. Influence of the time in culture on cellular and neuronal properties of neuroblastoma x glioma hybrid cells. With an appendix, mathematical description of the kinetics of the loss in cell volume. J Cell Sci. 1977;27:141–155. doi: 10.1242/jcs.27.1.141. [DOI] [PubMed] [Google Scholar]
- Hong MH, Xu C, Wang YJ, Ji JL, Tao YM, Xu XJ, Chen J, Xie X, Chi ZQ, Liu JG. Role of Src in ligand-specific regulation of delta-opioid receptor desensitization and internalization. J Neurochem. 2009;108:102–114. doi: 10.1111/j.1471-4159.2008.05740.x. [DOI] [PubMed] [Google Scholar]
- Hsia JA, Moss J, Hewlett EL, Vaughan M. ADP-ribosylation of adenylate cyclase by pertussis toxin. Effects on inhibitory agonist binding. J Biol Chem. 1984;259:1086–1090. [PubMed] [Google Scholar]
- Huang J, Lv Y, Fu Y, Ren L, Wang P, Liu B, Huang K, Bi J. Dynamic regulation of delta-opioid receptor in rat trigeminal ganglion neurons by lipopolysaccharide-induced acute pulpitis. J Endod. 2015 doi: 10.1016/j.joen.2015.09.011. [DOI] [PubMed] [Google Scholar]
- Hurley RW, Hammond DL. The analgesic effects of supraspinal mu and delta opioid receptor agonists are potentiated during persistent inflammation. J Neurosci. 2000;20:1249–1259. doi: 10.1523/JNEUROSCI.20-03-01249.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang L, Teng GM, Chan EY, Au SW, Wise H, Lee SS, Cheung WT. Impact of cell type and epitope tagging on heterologous expression of G protein-coupled receptor: a systematic study on angiotensin type II receptor. PLoS One. 2012;7:e47016. doi: 10.1371/journal.pone.0047016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones AK, Watabe H, Cunningham VJ, Jones T. Cerebral decreases in opioid receptor binding in patients with central neuropathic pain measured by [11C]diprenorphine binding and PET. Eur J Pain. 2004;8:479–485. doi: 10.1016/j.ejpain.2003.11.017. [DOI] [PubMed] [Google Scholar]
- Jutkiewicz EM, Baladi MG, Folk JE, Rice KC, Woods JH. The convulsive and electroencephalographic changes produced by nonpeptidic delta-opioid agonists in rats: comparison with pentylenetetrazol. J Pharmacol Exp Ther. 2006;317:1337–1348. doi: 10.1124/jpet.105.095810. [DOI] [PubMed] [Google Scholar]
- Kabli N, Cahill CM. Anti-allodynic effects of peripheral delta opioid receptors in neuropathic pain. Pain. 2007;127:84–93. doi: 10.1016/j.pain.2006.08.003. [DOI] [PubMed] [Google Scholar]
- Kazmi SM, Mishra RK. Opioid receptors in human neuroblastoma SH-SY5Y cells: evidence for distinct morphine (mu) and enkephalin (delta) binding sites. Biochem Biophys Res Commun. 1986;137:813–820. doi: 10.1016/0006-291x(86)91152-6. [DOI] [PubMed] [Google Scholar]
- Kazmi SM, Mishra RK. Comparative pharmacological properties and functional coupling of mu and delta opioid receptor sites in human neuroblastoma SH-SY5Y cells. Mol Pharmacol. 1987;32:109–118. [PubMed] [Google Scholar]
- Kieffer BL, Gaveriaux-Ruff C. Exploring the opioid system by gene knockout. Prog Neurobiol. 2002;66:285–306. doi: 10.1016/s0301-0082(02)00008-4. [DOI] [PubMed] [Google Scholar]
- Kieffer BL, Befort K, Gaveriaux-Ruff C, Hirth CG. The delta-opioid receptor: isolation of a cDNA by expression cloning and pharmacological characterization. Proc Natl Acad Sci U S A. 1992;89:12048–12052. doi: 10.1073/pnas.89.24.12048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klee WA, Nirenberg M. A neuroblastoma times glioma hybrid cell line with morphine receptors. Proc Natl Acad Sci U S A. 1974;71:3474–3477. doi: 10.1073/pnas.71.9.3474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klenowski P, Morgan M, Bartlett SE. The role of delta-opioid receptors in learning and memory underlying the development of addiction. Br J Pharmacol. 2015;172:297–310. doi: 10.1111/bph.12618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knapp RJ, Landsman R, Waite S, Malatynska E, Varga E, Haq W, Hruby VJ, Roeske WR, Nagase H, Yamamura HI. Properties of TAN-67, a nonpeptidic delta-opioid receptor agonist, at cloned human delta- and mu-opioid receptors. Eur J Pharmacol. 1995;291:129–134. doi: 10.1016/0922-4106(95)90134-5. [DOI] [PubMed] [Google Scholar]
- Kolb VM, Koman A, Terenius L. Fluorescent probes for opioid receptors. Life Sci. 1983;33(Suppl. 1):423–426. doi: 10.1016/0024-3205(83)90532-5. [DOI] [PubMed] [Google Scholar]
- Konig M, Zimmer AM, Steiner H, Holmes PV, Crawley JN, Brownstein MJ, Zimmer A. Pain responses, anxiety and aggression in mice deficient in pre-proenkephalin. Nature. 1996;383:535–538. doi: 10.1038/383535a0. [DOI] [PubMed] [Google Scholar]
- Kshirsagar T, Nakano AH, Law PY, Elde R, Portoghese PS. NTI4F: a non-peptide fluorescent probe selective for functional delta opioid receptors. Neurosci Lett. 1998;249:83–86. doi: 10.1016/s0304-3940(98)00379-6. [DOI] [PubMed] [Google Scholar]
- Lambert DG, Ghataorre AS, Nahorski SR. Muscarinic receptor binding characteristics of a human neuroblastoma SK-N-SH and its clones SH-SY5Y and SH-EP1. Eur J Pharmacol. 1989;165:71–77. doi: 10.1016/0014-2999(89)90771-1. [DOI] [PubMed] [Google Scholar]
- Law PY, Horn DS, Loh HH. Loss of opiate receptor activity in neuroblastoma X glioma NG108-15 hybrid cells after chronic opiate treatment. A multiple-step process. Mol Pharmacol. 1982;22:1–4. [PubMed] [Google Scholar]
- Law PY, Horn DS, Loh HH. Opiate receptor down-regulation and desensitization in neuroblastoma X glioma NG108-15 hybrid cells are two separate cellular adaptation processes. Mol Pharmacol. 1983;24:413–424. [PubMed] [Google Scholar]
- Le Merrer J, Becker JA, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev. 2009;89:1379–1412. doi: 10.1152/physrev.00005.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Merrer J, Plaza-Zabala A, Boca CD, Matifas A, Maldonado R, Kieffer BL. Deletion of the delta opioid receptor gene impairs place conditioning but preserves morphine reinforcement. Biol Psychiatry. 2011 doi: 10.1016/j.biopsych.2010.10.021. [DOI] [PubMed] [Google Scholar]
- Lee MC, Cahill CM, Vincent JP, Beaudet A. Internalization and trafficking of opioid receptor ligands in rat cortical neurons. Synapse. 2002;43:102–111. doi: 10.1002/syn.10014. [DOI] [PubMed] [Google Scholar]
- Lever JR, Scheffel U, Kinter CM, Ravert HT, Dannals RF, Wagner HN, Jr, Frost JJ. In vivo binding of N1′-([11C]methyl) naltrindole to delta-opioid receptors in mouse brain. Eur J Pharmacol. 1992;216:459–460. doi: 10.1016/0014-2999(92)90448-d. [DOI] [PubMed] [Google Scholar]
- Levitt ES, Purington LC, Traynor JR. Gi/o-coupled receptors compete for signaling to adenylyl cyclase in SH-SY5Y cells and reduce opioid-mediated cAMP overshoot. Mol Pharmacol. 2011;79:461–471. doi: 10.1124/mol.110.064816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma L, Cheng ZJ, Fan GH, Cai YC, Jiang LZ, Pei G. Functional expression, activation and desensitization of opioid receptor-like receptor ORL1 in neuroblastoma X glioma NG108-15 hybrid cells. FEBS Lett. 1997;403:91–94. doi: 10.1016/s0014-5793(97)00031-8. [DOI] [PubMed] [Google Scholar]
- Madar I, Bencherif B, Lever J, Heitmiller RF, Yang SC, Brock M, Brahmer J, Ravert H, Dannals R, Frost JJ. Imaging delta-and mu-opioid receptors by PET in lung carcinoma patients. J Nucl Med. 2007;48:207–213. [PubMed] [Google Scholar]
- Mansour A, Khachaturian H, Lewis ME, Akil H, Watson SJ. Anatomy of CNS opioid receptors. Trends Neurosci. 1988;11:308–314. doi: 10.1016/0166-2236(88)90093-8. [DOI] [PubMed] [Google Scholar]
- Marie N, Landemore G, Debout C, Jauzac P, Allouche S. Pharmacological characterization of AR-M1000390 at human delta opioid receptors. Life Sci. 2003a;73:1691–1704. doi: 10.1016/s0024-3205(03)00489-2. [DOI] [PubMed] [Google Scholar]
- Marie N, Lecoq I, Jauzac P, Allouche S. Differential sorting of human delta-opioid receptors after internalization by peptide and alkaloid agonists. J Biol Chem. 2003b;278:22795–22804. doi: 10.1074/jbc.M300084200. [DOI] [PubMed] [Google Scholar]
- Marie N, Aguila B, Hasbi A, Davis A, Jauzac P, Allouche S. Different kinases desensitize the human delta-opioid receptor (hDOP-R) in the neuroblastoma cell line SK-N-BE upon peptidic and alkaloid agonists. Cell Signal. 2008;20:1209–1220. doi: 10.1016/j.cellsig.2008.02.010. [DOI] [PubMed] [Google Scholar]
- Maslov LN, Lishmanov YB, Oeltgen PR, Barzakh El, Krylatov AV, Govindaswami M, Brown SA. Activation of peripheral delta2 opioid receptors increases cardiac tolerance to ischemia/reperfusion injury Involvement of protein kinase C, NO-synthase, KATP channels and the autonomic nervous system. Life Sci. 2009;84:657–663. doi: 10.1016/j.lfs.2009.02.016. [DOI] [PubMed] [Google Scholar]
- Matsuzawa S, Suzuki T, Misawa M, Nagase H. Involvement of mu- and delta-opioid receptors in the ethanol-associated place preference in rats exposed to foot shock stress. Brain Res. 1998;803:169–177. doi: 10.1016/s0006-8993(98)00679-9. [DOI] [PubMed] [Google Scholar]
- Mattia A, Vanderah T, Mosberg HI, Porreca F. Lack of antinociceptive cross-tolerance between [d-Pen2, d-Pen5] enkephalin and [d-Ala2]deltorphin II in mice: evidence for delta receptor subtypes. J Pharmacol Exp Ther. 1991;258:583–587. [PubMed] [Google Scholar]
- McDonald RL, Kaye DF, Reeve HL, Ball SG, Peers C, Vaughan PF. Bradykinin-evoked release of [3H]noradrenaline from the human neuroblastoma SH-SY5Y. Biochem Pharmacol. 1994;48:23–30. doi: 10.1016/0006-2952(94)90219-4. [DOI] [PubMed] [Google Scholar]
- McGinnity CJ, Shidahara M, Feldmann M, Keihaninejad S, Riano Barros DA, Gousias IS, Duncan JS, Brooks DJ, Heckemann RA, Turkheimer FE, Hammers A, Koepp MJ. Quantification of opioid receptor availability following spontaneous epileptic seizures: correction of [11C]diprenorphine PET data for the partial-volume effect. Neurolmage. 2013;79:72–80. doi: 10.1016/j.neuroimage.2013.04.015. [DOI] [PubMed] [Google Scholar]
- Melichar JK, Hume SP, Williams TM, Daglish MR, Taylor LG, Ahmad R, Malizia AL, Brooks DJ, Myles JS, Lingford-Hughes A, Nutt DJ. Using [11C]diprenorphine to image opioid receptor occupancy by methadone in opioid addiction: clinical and preclinical studies. J Pharmacol Exp Ther. 2005;312:309–315. doi: 10.1124/jpet.104.072686. [DOI] [PubMed] [Google Scholar]
- Mennicken F, Zhang J, Hoffert C, Ahmad S, Beaudet A, O’Donnell D. Phylogenetic changes in the expression of delta opioid receptors in spinal cord and dorsal root ganglia. J Comp Neural. 2003;465:349–360. doi: 10.1002/cne.10839. [DOI] [PubMed] [Google Scholar]
- Mika J, Popiolek-Barczyk K, Rojewska E, Makuch W, Starowicz K, Przewlocka B. Delta-opioid receptor analgesia is independent of microglial activation in a rat model of neuropathic pain. PLoS One. 2014;9:e104420. doi: 10.1371/journal.pone.0104420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monory K, Massa F, Egertova M, Eder M, Blaudzun H, Westenbroek R, Kelsch W, Jacob W, Marsch R, Ekker M, Long J, Rubenstein JL, Goebbels S, Nave KA, During M, Klugmann M, Wolfel B, Dodt HU, Zieglgansberger W, Wotjak CT, Mackie K, Elphick MR, Marsicano G, Lutz B. The endocannabinoid system controls key epileptogenic circuits in the hippocampus. Neuron. 2006;51:455–466. doi: 10.1016/j.neuron.2006.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morinville A, Cahill CM, Esdaile MJ, Aibak H, Collier B, Kieffer BL, Beaudet A. Regulation of delta-opioid receptor trafficking via mu-opioid receptor stimulation: evidence from mu-opioid receptor knock-out mice. J Neurosci. 2003;23:4888–4898. doi: 10.1523/JNEUROSCI.23-12-04888.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morinville A, Cahill CM, Aibak H, Rymar VV, Pradhan A, Hoffert C, Mennicken F, Stroh T, Sadikot AF, O’Donnell D, Clarke PB, Collier B, Henry JL, Vincent JP, Beaudet A. Morphine-induced changes in delta opioid receptor trafficking are linked to somatosensory processing in the rat spinal cord. J Neurosci. 2004;24:5549–5559. doi: 10.1523/JNEUROSCI.2719-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mosberg HI, Hurst R, Hruby VJ, Gee K, Yamamura HI, Galligan JJ, Burks TF. Bis-penicillamine enkephalins possess highly improved specificity toward delta opioid receptors. Proc Natl Acad Sci U S A. 1983;80:5871–5874. doi: 10.1073/pnas.80.19.5871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moses MA, Snell CR. The regulation of delta-opiate receptor density on 108CC15 neuroblastoma X glioma hybrid cells. Br J Pharmacol. 1984;81:169–174. doi: 10.1111/j.1476-5381.1984.tb10757.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nadal X, Banos JE, Kieffer BL, Maldonado R. Neuropathic pain is enhanced in delta-opioid receptor knockout mice. Eur J Neurosci. 2006;23:830–834. doi: 10.1111/j.1460-9568.2006.04569.x. [DOI] [PubMed] [Google Scholar]
- Nagase H, Yajima Y, Fujii H, Kawamura K, Narita M, Kamei J, Suzuki T. The pharmacological profile of delta opioid receptor ligands, (+) and (−) TAN-67 on pain modulation. Life Sci. 2001;68:2227–2231. doi: 10.1016/s0024-3205(01)01010-4. [DOI] [PubMed] [Google Scholar]
- Nagi K, Charfi I, Pineyro G. Kir3 channels undergo arrestin-dependant internalization following delta opioid receptor activation. Cell Mol Life Sci. 2015a doi: 10.1007/s00018-015-1899-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagi K, Charfi I, Pineyro G. Kir3 channels undergo arrestin-dependant internalization following delta opioid receptor activation. Cell Mol Life Sci. 2015b;72:3543–3557. doi: 10.1007/s00018-015-1899-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Negus SS, Gatch MB, Mello NK, Zhang X, Rice K. Behavioral effects of the delta-selective opioid agonist SNC80 and related compounds in rhesus monkeys. J Pharmacol Exp Ther. 1998;286:362–375. [PubMed] [Google Scholar]
- Negus SS, Rosenberg MB, Altarifi AA, O’Connell RH, Folk JE, Rice KC. Effects of the delta opioid receptor agonist SNC80 on pain-related depression of intracranial self-stimulation (ICSS) in rats. J Pain. 2012;13:317–327. doi: 10.1016/j.jpain.2011.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nowoczyn M, Marie N, Coulbault L, Hervault M, Davis A, Hanouz JL, Allouche S. Remifentanil produces cross-desensitization and tolerance with morphine on the mu-opioid receptor. Neuropharmacology. 2013;73:368–379. doi: 10.1016/j.neuropharm.2013.06.010. [DOI] [PubMed] [Google Scholar]
- Nozaki C, Nagase H, Nemoto T, Matifas A, Kieffer BL, Gaveriaux-Ruff C. In vivo properties of KNT-127, a novel delta opioid agonist: receptor internalisation, antihyperalgesia and antidepressant effects in mice. Br J Pharmacol. 2014 doi: 10.1111/bph.12852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otis V, Sarret P, Gendron L. Spinal activation of delta opioid receptors alleviates cancer-related bone pain. Neuroscience. 2011;183:221–229. doi: 10.1016/j.neuroscience.2011.03.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Overland AC, Kitto KF, Chabot-Dore AJ, Rothwell PE, Fairbanks CA, Stone LS, Wilcox GL. Protein kinase C mediates the synergistic interaction between agonists acting at alpha2-adrenergic and delta-opioid receptors in spinal cord. J Neurosci. 2009;29:13264–13273. doi: 10.1523/JNEUROSCI.1907-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palazzi E, Ceppi E, Guglielmetti F, Catozzi L, Amoroso D, Groppetti A. Biochemical evidence of functional interaction between mu- and delta-opioid receptors in SK-N-BE neuroblastoma cell line. J Neurochem. 1996;67:138–144. doi: 10.1046/j.1471-4159.1996.67010138.x. [DOI] [PubMed] [Google Scholar]
- Pasquini F, Bochet P, Garbay-Jaureguiberry C, Roques BP, Rossier J, Beaudet A. Electron microscopic localization of photoaffinity-labelled delta opioid receptors in the neostriatum of the rat. J Comp Neurol. 1992;326:229–244. doi: 10.1002/cne.903260206. [DOI] [PubMed] [Google Scholar]
- Pasquini F, Jomary C, Garbay-Jaureguiberry C, Roques BP, Beaudet A. [125l]azido-DTLET as a tool for selective covalent labeling of delta-opioid receptors in rat brain sections. Eur J Pharmacol. 1993;243:39–45. doi: 10.1016/0014-2999(93)90165-e. [DOI] [PubMed] [Google Scholar]
- Payza K. Binding and activity of opioid ligands at the cloned human delta, mu, and kappa receptors. In: Chang KJ, Porrecca F, Woods JH, editors. The Delta Receptor. New York: Marcel Dekker, Inc; 2004. pp. 216–275. [Google Scholar]
- Perrine SA, Hoshaw BA, Unterwald EM. Delta opioid receptor ligands modulate anxiety-like behaviors in the rat. Br J Pharmacol. 2006;147:864–872. doi: 10.1038/sj.bjp.0706686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Platika D, Boulos MH, Baizer L, Fishman MC. Neuronal traits of clonal cell lines derived by fusion of dorsal root ganglia neurons with neuroblastoma cells. Proc Natl Acad Sci U S A. 1985;82:3499–3503. doi: 10.1073/pnas.82.10.3499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polastron J, Mur M, Mazarguil H, Puget A, Meunier JC, Jauzac P. SK-N-BE: a human neuroblastoma cell line containing two subtypes of delta-opioid receptors. J Neurochem. 1994;62:898–906. doi: 10.1046/j.1471-4159.1994.62030898.x. [DOI] [PubMed] [Google Scholar]
- Poole DP, Pelayo JC, Scherrer G, Evans CJ, Kieffer BL, Bunnett NW. Localization and regulation of fluorescently labeled delta opioid receptor, expressed in enteric neurons of mice. Gastroenterology. 2011;141:982–991. doi: 10.1053/j.gastro.2011.05.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Portoghese PS, Sultana M, Takemori AE. Naltrindole, a highly selective and potent non-peptide delta opioid receptor antagonist. Eur J Pharmacol. 1988;146:185–186. doi: 10.1016/0014-2999(88)90502-x. [DOI] [PubMed] [Google Scholar]
- Pradhan AA, Clarke PB. Comparison between delta-opioid receptor functional response and autoradiographic labeling in rat brain and spinal cord. J Comp Neurol. 2005a;481:416–426. doi: 10.1002/cne.20378. [DOI] [PubMed] [Google Scholar]
- Pradhan AA, Clarke PB. Comparison between delta-opioid receptor functional response and autoradiographic labeling in rat brain and spinal cord. J Comp Neurol. 2005b;481:416–426. doi: 10.1002/cne.20378. [DOI] [PubMed] [Google Scholar]
- Pradhan AA, Becker JA, Scherrer G, Tryoen-Toth P, Filliol D, Matifas A, Massotte D, Gaveriaux-Ruff C, Kieffer BL. In vivo delta opioid receptor internalization controls behavioral effects of agonists. PLoS One. 2009;4:e5425. doi: 10.1371/journal.pone.0005425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Walwyn W, Nozaki C, Filliol D, Erbs E, Matifas A, Evans C, Kieffer BL. Ligand-directed trafficking of the delta-opioid receptor in vivo: two paths toward analgesic tolerance. J Neurosci. 2010;30:16459–16468. doi: 10.1523/JNEUROSCI.3748-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Befort K, Nozaki C, Gaveriaux-Ruff C, Kieffer BL. The delta opioid receptor: an evolving target for the treatment of brain disorders. Trends Pharmacol Sci. 2011a doi: 10.1016/j.tips.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Befort K, Nozaki C, Gaveriaux-Ruff C, Kieffer BL. The delta opioid receptor: an evolving target for the treatment of brain disorders. Trends Pharmacol Sci. 2011b;32:581–590. doi: 10.1016/j.tips.2011.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Smith ML, Kieffer BL, Evans CJ. Ligand-directed signalling within the opioid receptor family. Br J Pharmacol. 2012;167:960–969. doi: 10.1111/j.1476-5381.2012.02075.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan A, Smith M, McGuire B, Evans C, Walwyn W. Chronic inflammatory injury results in increased coupling of delta opioid receptors to voltage-gated Ca2+ channels. Molecular pain. 2013;9:8. doi: 10.1186/1744-8069-9-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Smith ML, Zyuzin J, Charles A. Delta-Opioid receptor agonists inhibit migraine-related hyperalgesia, aversive state and cortical spreading depression in mice. Br J Pharmacol. 2014;171:2375–2384. doi: 10.1111/bph.12591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Tawfik VL, Tipton AF, Scherrer G. In vivo techniques to investigate the internalization profile of opioid receptors. Methods Mol Biol. 2015;1230:87–104. doi: 10.1007/978-1-4939-1708-2_7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pradhan AA, Perroy J, Walwyn WM, Smith ML, Vicente-Sanchez A, Segura L, Bana A, Kieffer BL, Evans CJ. Agonist-Specific Recruitment of Arrestin Isoforms Differentially Modify Delta Opioid Receptor Function. J Neurosci. 2016;36:3541–3551. doi: 10.1523/JNEUROSCI.4124-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puthenveedu MA, Lauffer B, Temkin P, Vistein R, Carlton P, Thorn K, Taunton J, Weiner OD, Parton RG, von Zastrow M. Sequence-dependent sorting of recycling proteins by actin-stabilized endosomal microdomains. Cell. 2010;143:761–773. doi: 10.1016/j.cell.2010.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu Y, Loh HH, Law PY. Phosphorylation of the delta-opioid receptor regulates its beta-arrestins selectivity and subsequent receptor internalization and adenylyl cyclase desensitization. J Biol Chem. 2007;282:22315–22323. doi: 10.1074/jbc.M611258200. [DOI] [PubMed] [Google Scholar]
- Richard-Lalonde M, Nagi K, Audet N, Sleno R, Amraei M, Hogue M, Balboni G, Schiller PW, Bouvier M, Hebert TE, Pineyro G. Conformational dynamics of Kir3.1/Kir3.2 channel activation via delta-opioid receptors. Mol Pharmacol. 2013;83:416–428. doi: 10.1124/mol.112.081950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richards EM, Mathews DC, Luckenbaugh DA, lonescu DF, Machado-Vieira R, Niciu MJ, Duncan WC, Nolan NM, Franco-Chaves JA, Hudzik T, Maciag C, Li S, Cross A, Smith MA, Zarate CA., Jr A randomized, placebo-controlled pilot trial of the delta opioid receptor agonist AZD2327 in anxious depression. Psychopharmacology. 2016 doi: 10.1007/s00213-015-4195-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riedl MS, Schnell SA, Overland AC, Chabot-Dore AJ, Taylor AM, Ribeiro-da-Silva A, Elde RP, Wilcox GL, Stone LS. Coexpression of alpha 2A–adrenergic and delta-opioid receptors in substance P-containing terminals in rat dorsal horn. J Comp Neurol. 2009;513:385–398. doi: 10.1002/cne.21982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rochon K, Proteau-Gagne A, Bourassa P, Nadon JF, Cote J, Bournival V, Gobeil F, Jr, Guerin B, Dory YL, Gendron L. Preparation and evaluation at the delta opioid receptor of a series of linear leuenkephalin analogues obtained by systematic replacement of the amides. ACS Chem Neurosci. 2013;4:1204–1216. doi: 10.1021/cn4000583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roerig SC, Loh HH, Law PY. Identification of three separate guanine nucleotide-binding proteins that interact with the delta-opioid receptor in NG108-15 neuroblastoma x glioma hybrid cells. Mol Pharmacol. 1992;41:822–831. [PubMed] [Google Scholar]
- Rubovitch V, Gafni M, Same Y. The mu opioid agonist DAMGO stimulates cAMP production in SK-N-SH cells through a PLC-PKC-Ca + + pathway. Brain Res Mol Brain Res. 2003;110:261–266. doi: 10.1016/s0169-328x(02)00656-3. [DOI] [PubMed] [Google Scholar]
- Ruest LB, Hammer RE, Yanagisawa M, Clouthier DE. Dlx5/6-enhancer directed expression of Cre recombinase in the pharyngeal arches and brain. Genesis. 2003;37:188–194. doi: 10.1002/gene.10247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saitoh A, Kimura Y, Suzuki T, Kawai K, Nagase H, Kamei J. Potential anxiolytic and antidepressant-like activities of SNC80, a selective delta-opioid agonist, in behavioral models in rodents. J Pharmacol Sci. 2004;95:374–380. doi: 10.1254/jphs.fpj04014x. [DOI] [PubMed] [Google Scholar]
- Saitoh A, Yoshikawa Y, Onodera K, Kamei J. Role of delta-opioid receptor subtypes in anxiety-related behaviors in the elevated plus-maze in rats. Psychopharmacology. 2005;182:327–334. doi: 10.1007/s00213-005-0112-6. [DOI] [PubMed] [Google Scholar]
- Sanchez-Blazquez P, Garzon J. Mastoparan reduces the supraspinal analgesia mediated by mu/delta-opioid receptors in mice. Eur J Pharmacol. 1994;258:159–162. doi: 10.1016/0014-2999(94)90070-1. [DOI] [PubMed] [Google Scholar]
- Same Y, Rubovitch V, Fields A, Gafni M. Dissociation between the inhibitory and stimulatory effects of opioid peptides on cAMP formation in SK-N-SH neuroblastoma cells. Biochem Biophys Res Commun. 1998;246:128–131. doi: 10.1006/bbrc.1998.8582. [DOI] [PubMed] [Google Scholar]
- Scherrer G, Befort K, Contet C, Becker J, Matifas A, Kieffer BL. The delta agonists DPDPE and deltorphin II recruit predominantly mu receptors to produce thermal analgesia: a parallel study of mu, delta and combinatorial opioid receptor knockout mice. Eur J Neurosci. 2004;19:2239–2248. doi: 10.1111/j.0953-816X.2004.03339.x. [DOI] [PubMed] [Google Scholar]
- Scherrer G, Tryoen-Toth P, Filliol D, Matifas A, Laustriat D, Cao YQ, Basbaum Al, Dierich A, Vonesh JL, Gaveriaux-Ruff C, Kieffer BL. Knockin mice expressing fluorescent delta-opioid receptors uncover G protein-coupled receptor dynamics in vivo. Proc Natl Acad Sci U S A. 2006;103:9691–9696. doi: 10.1073/pnas.0603359103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scherrer G, Imamachi N, Cao YQ, Contet C, Mennicken F, O’Donnell D, Kieffer BL, Basbaum Al. Dissociation of the opioid receptor mechanisms that control mechanical and heat pain. Cell. 2009;137:1148–1159. doi: 10.1016/j.cell.2009.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schiller PW, Nguyen TM, Weltrowska G, Wilkes BC, Marsden BJ, Lemieux C, Chung NN. Differential stereochemical requirements of mu vs. delta opioid receptors for ligand binding and signal transduction: development of a class of potent and highly delta-selective peptide antagonists. Proc Natl Acad Sci U S A. 1992a;89:11871–11875. doi: 10.1073/pnas.89.24.11871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schiller PW, Weltrowska G, Nguyen TM, Wilkes BC, Chung NN, Lemieux C. Conformationally restricted deltorphin analogues. J Med Chem. 1992b;35:3956–3961. doi: 10.1021/jm00099a025. [DOI] [PubMed] [Google Scholar]
- Schiller PW, Weltrowska G, Nguyen TM, Wilkes BC, Chung NN, Lemieux C. TIPP[psi]: a highly potent and stable pseudopeptide delta opioid receptor antagonist with extraordinary delta selectivity. J Med Chem. 1993;36:3182–3187. doi: 10.1021/jm00073a020. [DOI] [PubMed] [Google Scholar]
- Schuster DJ, Metcalf MD, Kitto KF, Messing RO, Fairbanks CA, Wilcox GL. Ligand requirements for involvement of PKC epsilon in synergistic analgesic interactions between spinal mu and delta opioid receptors. Br J Pharmacol. 2015;172:642–653. doi: 10.1111/bph.12774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shields SD, Ahn HS, Yang Y, Han C, Seal RP, Wood JN, Waxman SG, Dib-Hajj SD. Nav1.8 expression is not restricted to nociceptors in mouse peripheral nervous system. Pain. 2012;153:2017–2030. doi: 10.1016/j.pain.2012.04.022. [DOI] [PubMed] [Google Scholar]
- Sofuoglu M, Portoghese PS, Takemori AE. Cross-tolerance studies in the spinal cord of beta-FNA-treated mice provides further evidence for delta opioid receptor subtypes. Life Sci. 1991;49:PL153–PL156. doi: 10.1016/0024-3205(91)90397-t. [DOI] [PubMed] [Google Scholar]
- Stevenson GW, Folk JE, Rice KC, Negus SS. Interactions between delta and mu opioid agonists in assays of schedule-controlled responding, thermal nociception, drug self-administration, and drug versus food choice in rhesus monkeys: studies with SNC80 [(+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenz yl]-N,N-diethylbenzamide] and heroin. J Pharmacol Exp Ther. 2005;314:221–231. doi: 10.1124/jpet.104.082685. [DOI] [PubMed] [Google Scholar]
- Sugiyama A, Nagase H, Oka J, Yamada M, Saitoh A. DOR(2)-selective but not DOR(1)-selective antagonist abolishes anxiolytic-like effects of the delta opioid receptor agonist KNT-127. Neuropharmacology. 2014;79:314–320. doi: 10.1016/j.neuropharm.2013.11.021. [DOI] [PubMed] [Google Scholar]
- Suzuki T, Tsuji M, Mori T, Misawa M, Endoh T, Nagase H. Effects of a highly selective nonpeptide delta opioid receptor agonist, TAN-67, on morphine-induced antinociception in mice. Life Sci. 1995;57:155–168. doi: 10.1016/0024-3205(95)00256-6. [DOI] [PubMed] [Google Scholar]
- Svingos AL, Clarke CL, Pickel VM. Cellular sites for activation of delta-opioid receptors in the rat nucleus accumbens shell: relationship with Met5-enkephalin. J Neurosci. 1998;18:1923–1933. doi: 10.1523/JNEUROSCI.18-05-01923.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tao PL, Han KF, Wang SD, Lue WM, Elde R, Law PY, Loh HH. Immunohistochemical evidence of down-regulation of mu-opioid receptor after chronic PL-017 in rats. Eur J Pharmacol. 1998;344:137–142. doi: 10.1016/s0014-2999(97)01596-3. [DOI] [PubMed] [Google Scholar]
- Tseng LF, Narita M, Mizoguchi H, Kawai K, Mizusuna A, Kamei J, Suzuki T, Nagase H. Delta-1 opioid receptor-mediated antinociceptive properties of a nonpeptidic delta opioid receptor agonist, (−)TAN-67, in the mouse spinal cord. J Pharmacol Exp Ther. 1997;280:600–605. [PubMed] [Google Scholar]
- Tudashki HB, Robertson DN, Schiller PW, Pineyro G. Endocytic profiles of delta-opioid receptor ligands determine the duration of rapid but not sustained cAMP responses. Mol Pharmacol. 2014;85:148–161. doi: 10.1124/mol.113.089003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyacke RJ, Robinson ES, Schnabel R, Lewis JW, Husbands SM, Nutt DJ, Hudson AL. N1′-fluoroethyl-naltrindole (BU97001) and N1′-fluoroethyl-(14-formylamino)-naltrindole (BU97018) potential delta-opioid receptor PET ligands. Nucl Med Biol. 2002;29:455–462. doi: 10.1016/s0969-8051(02)00300-1. [DOI] [PubMed] [Google Scholar]
- van Rijn RM, Whistler JL. The delta(1) opioid receptor is a heterodimer that opposes the actions of the delta(2) receptor on alcohol intake. Biol Psychiatry. 2009a;66:777–784. doi: 10.1016/j.biopsych.2009.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Rijn RM, Whistler JL. The delta(1) opioid receptor is a heterodimer that opposes the actions of the delta(2) receptor on alcohol intake. Biol Psychiatry. 2009b;66:777–784. doi: 10.1016/j.biopsych.2009.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Rijn RM, Brissett Dl, Whistler JL. Dual efficacy of delta opioid receptor-selective ligands for ethanol drinking and anxiety. J Pharmacol Exp Ther. 2010;335:133–139. doi: 10.1124/jpet.110.170969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Rijn RM, Brissett Dl, Whistler JL. Emergence of functional spinal delta opioid receptors after chronic ethanol exposure. Biol Psychiatry. 2012;71:232–238. doi: 10.1016/j.biopsych.2011.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Rijn RM, Defriel JN, Whistler JL. Pharmacological traits of delta opioid receptors: pitfalls or opportunities? Psychopharmacology. 2013;228:1–18. doi: 10.1007/s00213-013-3129-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wand GS, Weerts EM, Kuwabara H, Wong DF, Xu X, McCaul ME. The relationship between naloxone-induced cortisol and delta opioid receptor availability in mesolimbic structures is disrupted in alcohol-dependent subjects. Addiction biology. 2013;18:181–192. doi: 10.1111/j.1369-1600.2011.00430.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Pickel VM. Preferential cytoplasmic localization of delta-opioid receptors in rat striatal patches: comparison with plasmalemmal mu-opioid receptors. J Neurosci. 2001;21:3242–3250. doi: 10.1523/JNEUROSCI.21-09-03242.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Traynor JR. Opioid-induced down-regulation of RGS4: role of ubiquitination and implications for receptor crosstalk. J Biol Chem. 2011;286:7854–7864. doi: 10.1074/jbc.M110.160911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang HB, Guan JS, Bao L, Zhang X. Distinct subcellular distribution of delta-opioid receptor fused with various tags in PC12 cells. Neurochem Res. 2008;33:2028–2034. doi: 10.1007/s11064-008-9678-9. [DOI] [PubMed] [Google Scholar]
- Wang Q, Liu-Chen LY, Traynor JR. Differential modulation of mu- and delta-opioid receptor agonists by endogenous RGS4 protein in SH-SY5Y cells. J Biol Chem. 2009;284:18357–18367. doi: 10.1074/jbc.M109.015453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang HB, Zhao B, Zhong YQ, Li KC, Li ZY, Wang Q, Lu YJ, Zhang ZN, He SQ, Zheng HC, Wu SX, Hokfelt TG, Bao L, Zhang X. Coexpression of delta- and mu-opioid receptors in nociceptive sensory neurons. Proc Natl Acad Sci U S A. 2010;107:13117–13122. doi: 10.1073/pnas.1008382107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weerts EM, Kim YK, Wand GS, Dannals RF, Lee JS, Frost JJ, McCaul ME. Differences in delta- and mu-opioid receptor blockade measured by positron emission tomography in naltrexone-treated recently abstinent alcohol-dependent subjects. Neuropsychopharmacology. 2008;33:653–665. doi: 10.1038/sj.npp.1301440. [DOI] [PubMed] [Google Scholar]
- Weerts EM, Wand GS, Kuwabara H, Munro CA, Dannals RF, Hilton J, Frost JJ, McCaul ME. Positron emission tomography imaging of mu- and delta-opioid receptor binding in alcohol-dependent and healthy control subjects. Alcohol Clin Exp Res. 2011;35:2162–2173. doi: 10.1111/j.1530-0277.2011.01565.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei ZY, Brown W, Takasaki B, Plobeck N, Delorme D, Zhou F, Yang H, Jones P, Gawell L, Gagnon H, Schmidt R, Yue SY, Walpole C, Payza K, St Onge S, Labarre M, Godbout C, Jakob A, Butterworth J, Kamassah A, Morin PE, Projean D, Ducharme J, Roberts E. N,N-Diethyl-4-(phenylpiperidin-4-ylidenemethyl) benzamide: a novel, exceptionally selective, potent delta opioid receptor agonist with oral bioavailability and its analogues. J Med Chem. 2000;43:3895–3905. doi: 10.1021/jm000229p. [DOI] [PubMed] [Google Scholar]
- Whistler JL, Enquist J, Marley A, Fong J, Gladher F, Tsuruda P, Murray SR, von Zastrow M. Modulation of postendocytic sorting of G protein-coupled receptors. Science. 2002;297:615–620. doi: 10.1126/science.1073308. [DOI] [PubMed] [Google Scholar]
- Williams SA, Abbruscato TJ, Hruby VJ, Davis TP. Passage of a delta-opioid receptor selective enkephalin, [d-penicillamine2,5] enkephalin, across the blood-brain and the blood-cerebrospinal fluid barriers. J Neurochem. 1996;66:1289–1299. doi: 10.1046/j.1471-4159.1996.66031289.x. [DOI] [PubMed] [Google Scholar]
- Williams TM, Davies SJ, Taylor LG, Daglish MR, Hammers A, Brooks DJ, Nutt DJ, Lingford-Hughes A. Brain opioid receptor binding in early abstinence from alcohol dependence and relationship to craving: an [11C]diprenorphine PET study. Eur Neuropsychopharmacol. 2009;19:740–748. doi: 10.1016/j.euroneuro.2009.06.007. [DOI] [PubMed] [Google Scholar]
- Willoch F, Schindler F, Wester HJ, Empl M, Straube A, Schwaiger M, Conrad B, Tolle TR. Central poststroke pain and reduced opioid receptor binding within pain processing circuitries: a [11C] diprenorphine PET study. Pain. 2004;108:213–220. doi: 10.1016/j.pain.2003.08.014. [DOI] [PubMed] [Google Scholar]
- Yalcin I, Barrot M. The anxiodepressive comorbidity in chronic pain. Curr Opin Anaesthesiol. 2014;27:520–527. doi: 10.1097/ACO.0000000000000116. [DOI] [PubMed] [Google Scholar]
- Yu VC, Sadee W. Efficacy and tolerance of narcotic analgesics at the mu opioid receptor in differentiated human neuroblastoma cells. J Pharmacol Exp Ther. 1988;245:350–355. [PubMed] [Google Scholar]
- Yu VC, Richards ML, Sadee W. A human neuroblastoma cell line expresses mu and delta opioid receptor sites. J Biol Chem. 1986;261:1065–1070. [PubMed] [Google Scholar]
- Zajac JM, Rostene W, Roques BP. Irreversible labelling of delta-opioid receptors in rat brain and neuroblastoma cells by [3H] azido-DTLET: characterization of subunits and autoradiographic visualization of the covalent binding. Neuropeptides. 1987;9:295–307. doi: 10.1016/0143-4179(87)90004-7. [DOI] [PubMed] [Google Scholar]
- Zhang X, Bao L, Guan JS. Role of delivery and trafficking of delta-opioid peptide receptors in opioid analgesia and tolerance. Trends Pharmacol Sci. 2006;27:324–329. doi: 10.1016/j.tips.2006.04.005. [DOI] [PubMed] [Google Scholar]
- Zhang X, Bao L, Li S. Opioid receptor trafficking and interaction in nociceptors. Br J Pharmacol. 2015;172:364–374. doi: 10.1111/bph.12653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu Y, King MA, Schuller AG, Nitsche JF, Reidl M, Elde RP, Unterwald E, Pasternak GW, Pintar JE. Retention of supraspinal delta-like analgesia and loss of morphine tolerance in delta opioid receptor knockout mice. Neuron. 1999;24:243–252. doi: 10.1016/s0896-6273(00)80836-3. [DOI] [PubMed] [Google Scholar]