Abstract
Nalfurafine, a moderately selective kappa opioid receptor (KOR) agonist, is used in Japan for treatment of itch without causing dysphoria or psychotomimesis. Here we characterized pharmacology of compound 42B, a 3-dehydroxy analog of nalfurafine and compared with that of nalfurafine. Nalfurafine and 42B acted as full KOR agonists and partial μ opioid receptor (MOR) agonists, but 42B showed much lower potency for both receptors and lower KOR/MOR selectivity, different from previous reports. Molecular modeling revealed that water-mediated hydrogen-bond formation between 3-OH of nalfurafine, and KOR accounted for its higher KOR potency than 42B. The higher potency of both at KOR over MOR may be due to hydrogen-bond formation between non-conserved Y7.35 of KOR and their carbonyl groups. Both showed modest G protein signaling biases. In mice, like nalfurafine, 42B produced antinociceptive and anti-scratch effects and did not cause conditioned place aversion (CPA) in the effective dose ranges. Unlike nalfurafine, 42B caused motor incoordination and hypolocomotion. As both agonists showed G protein biases, yet produced different effects on locomotor activity and motor incoordination, the findings and those in the literature suggest caution in correlating in vitro biochemical data with in vivo behavior effects. The factors contributing to the disconnect, including pharmacodynamic and pharmacokinetic issues, are discussed. In addition, our results suggest that among the KOR-induced adverse behaviors, CPA can be separated from motor incoordination and hypolocomotion.
Keywords: kappa opioid receptor, nalfurafine, antipruritic effect, conditioned place aversion, sedation, motor incoordination
Introduction
The κ opioid receptor (KOR), one of the three opioid receptors, belongs to the rhodopsin subfamily of G protein-coupled receptors. KOR agonists produce analgesic and antipruritic effects1–4; however, clinical development of these compounds has been limited by the side effects such as psychotomimetic effect, dysphoria, and sedation2, 4, 5, except for nalfurafine. Nalfurafine, a moderately selective KOR agonist, has been used in Japan for treatment of itch in patients undergoing hemodialysis and those with chronic liver diseases6–10. Clinical reports have shown that different from other KOR agonists that have been tested in humans, at therapeutic doses, nalfurafine did not produce dysphoria or psychotomimetic effects7–9. Recently, we have demonstrated that in mice nalfurafine exerted analgesic and anti-scratch effects without adverse effects such as sedation, motor incoordination, or conditioned place aversion (CPA)11, thus recapitulating the observations in humans. Because of its favorable side effect profile, nalfurafine was also investigated as an adjuvant to μ opioid receptor (MOR)-targeting analgesics12 and as a therapeutic agent for drug abuse and alcohol treatment13–16. Nalfurafine enhanced analgesic effects of morphine, but reduced morphine-induced conditioned place preference (CPP) and hyperlocomotion in mice12. Preclinical research showed that nalfurafine augmented antinociceptive effect of oxycodone, but attenuated oxycodone-induced reinforcing effects and respiratory depression13, 14. In addition, nalfurafine reduced alcohol intake and relapse-like alcohol drinking and repeated administration did not induce tolerance15, 16.
Although nalfurafine is a full agonist for the KOR and a partial agonist for the MOR in [35S]GTPγS binding assays17, its analgesic and anti-scratching effects were shown to be mediated by the KOR18–22. Its effects on reducing drug abuse was also mediated by KOR since oxycodone self-administration and alcohol drinking were blocked by the selective KOR antagonist nor-binaltorphimine (norBNI)13–16. Nalfurafine did not cause CPA in MOR knockout mice, indicating that its lack of CPA in wildtype mice is not due to its combined actions on the MOR and KOR11. Thus, selective KOR agonists with similar chemical structures as nalfurafine may be promising therapeutic agents with fewer side effects.
42B, a nalfurafine analog which lacks the 3-hydroxy moiety of the phenolic group (Figure 1 a,b), was reported to show higher selectivity for KOR over MOR than nalfurafine, albeit with lower affinities for both receptors in radioligand binding assays23, 24. However, no in vivo characterization was performed on 42B. We hypothesized that 42B may show favorable pharmacological properties in vivo due to its similar chemical structure to nalfurafine and higher selectivity for KOR than nalfurafine. Although the 3-OH group of morphinan opioids was described long ago as one of the most important pharmacophore groups, such a conclusion mainly came from the small molecule structure-activity relationship with very limited information from structural biology due to technical limitation previously. Recently, as all the three major opioid receptors have been crystallized in both active and inactive states, different conclusions have been reported on the role of 3-OH group for its influence on binding affinity and selectivity for the KOR activated structure24. It was suggested that modifications of the phenol group may be a possible path towards producing selective KOR ligands with desirable pharmacological characteristics24. Thus, a revisit of many take-for-granted concepts and opinions in this field is warranted. Therefore, in this study, we examined in vitro and in vivo pharmacological properties of the 3-dehydroxy analog of nalfurafine, 42B, and compared with those of nalfurafine.
Figure 1.
The chemical structures of nalfurafine (a) and 42B (b) with atom notations.
Results and Discussion
Maximal responses and potencies of nalfurafine and 42B in stimulating [35S]GTPγS binding via MOR, δ opioid receptor (DOR), KOR and nociceptin / orphanin FQ receptor (NOR).
[35S]GTPγS binding was applied as a functional measure of activation of opioid receptors and NOR. Nalfurafine and its 3-dehydroxy analog 42B stimulated the KOR to enhance [35S]GTPγS binding to membranes, both having the maximal effects 91% of that of the full agonist (−)U50,488 (Figure 2 a–d; Table 1), indicating that both are full agonists at the KOR in this assay. The potency of nalfurafine (EC50 = 0.097 ± 0.018 nM) at the KOR was ~250-fold higher than that of 42B (EC50 = 25.56 ± 1.50 nM).
Figure 2. Stimulation of [35S]GTPγS binding to membranes of CHO cells stably transfected with the MOR, DOR, KOR or NOR by nalfurafine, 42B, and a reference full agonist for each receptor.
[35S]GTPγS binding to membranes was performed with various concentrations of each compound in 50 mM Tris buffer (pH 7.4) in the presence of 50 mM (for the MOR, DOR or NOR) or 100 mM NaCl (for the KOR), 5 mM MgCl2, 15 μM GDP, 1 mM EDTA with ~10 μg membrane proteins and ~80 pM [35S]GTPγS at 30°C for 60 min. Nonspecific binding, determined using 10 μM cold GTPγS, was ~500 dpm. Data were normalized and expressed as percentage of the maximal [35S]GTPγS binding of the respective reference full agonist. Basal [35S]GTPγS binding in the absence of added compounds was ~2000 dpm and maximal binding (with basal binding subtracted) was ~2500 dpm for the MOR and DOR, ~5000 dpm for the KOR, ~1500 dpm for the NOR. Each value represents the mean ± SEM of at least three independent experiments performed in duplicate. EC50 values and maximal responses are shown in Table 1. Each value is mean ± SEM (n=3–4).
Table 1.
EC50 values and maximal effects of nalfurafine and 42B in stimulating [35S]GTPγS binding to membranes of CHO cells stably expressing the MOR, DOR, KOR or NOR. DAMGO, DPDPE, (−)U50,488H, and N/OFQ were used as the reference full agonists for the MOR, DOR, KOR and NOR, respectively. Data were derived from the curves in Figure 2. Mean ± SEM (n=3–4)
MOR | DOR | KOR | NOR | |||||
---|---|---|---|---|---|---|---|---|
EC50 | Maximal Effect | EC50 | Maximal Effect | EC50 | Maximal Effect | EC50 | Maximal Effect | |
nM | % | nM | % | nM | % | nM | % | |
DAMGO | 6.30 ± 0.43 | 99.42 ± 1.07 | ||||||
DPDPE | 7.06 ± 0.76 | 97.67 ± 1.92 | ||||||
(−)U50,488H | 5.12 ± 0.37 | 99.65 ± 1.24 | ||||||
Nociceptin / OFQ | 0.046 ± 0.007 | 110.8 ± 2.8 | ||||||
Nalfurafine | 3.11 ± 0.63 | 73.88 ± 2.93 | 24.22 ± 2.56 | 129.1 ± 3.2 | 0.097 ± 0.018 | 90.90 ± 3.25 | 279.7 ± 17.7 | 119.7 ± 2.2 |
42B | 214.9 ± 50.4 | 48.87 ± 2.45 | 334.5 ± 35.1 | 134.7 ± 3.4 | 25.56 ± 1.50 | 91.34 ± 1.15 | 1760 ± 284 | 110.9 ± 7.4 |
For the MOR, both nalfurafine and 42B acted as partial agonists, with the maximal effect being 74% and 49%, respectively, of that of the full agonist DAMGO. Nalfurafine also showed higher potency at the MOR than 42B, with the EC50 values of 3.11 ± 0.63 nM and 214.9 ± 50.4 nM, respectively. Thus, nalfurafine not only showed higher potency in activating both the KOR and the MOR, but also had higher selectivity for the KOR over the MOR (32-fold) than 42B (8.4-fold).
In addition, nalfurafine and 42B demonstrated full agonism at the DOR and NOR with relatively low potency.
Our results that nalfurafine showed greater selectivity for the KOR over the MOR than 42B in [35S]GTPγS binding assays are different from the previous radioligand binding results that nalfurafine showed lower ratio of KOR/MOR affinity than 42B23, 24. Nagase and Fujii23 reported that the ratios of KOR/MOR affinity were 2.4 for nalfurafine and 60 for 42B (Supplementary Materials, Table S1). Che, et al.24 showed that the KOR/MOR affinity ratios of nalfurafine and 42B (compound 18) were 13.1 and 355, respectively (Table S1). Assays and tissues used may account for the discrepancy. We performed [35S]GTPγS binding, a functional measure of receptor activation, whereas both Nagase and Fujii23 and Che et al.24 did inhibition of radioligand binding. As shown in Table S1, Che et al.24 did inhibition of [3H]diprenorphine binding to the KOR and MOR stably expressed in Sf9 cells. Nagase and Fujii23 determined the affinities by inhibition of [3H]U69,593 binding to the KOR in guinea pig cerebellum membranes and of [3H]DAMGO binding to the MOR in mouse brain membranes.
Effects of nalfurafine and 42B on KOR-mediated G protein and β-arrestin signaling
For determination of the effects of nalfurafine and 42B on G protein- and β-arrestin-mediated signaling following KOR activation, GloSensor cAMP assay and Tango assay were performed by the Psychoactive Drug Screening Program (PDSP) of the National Institute of Mental Health (NIMH) for G protein activation and β-arrestin2 recruitment, respectively. The prototypic KOR agonist U50,488H was used as the reference agonist. Nalfurafine and 42B activated G proteins and recruited β-arrestin in a dose-dependent fashion with high maximal responses and varying potencies in the two assays (Figure 3, Table 2). For inhibition of cAMP accumulation, both nalfurafine and 42B had nearly maximal effects as U50,488H, but nalfurafine (EC50 = 0.17 ± 0.04 nM) and 42B (EC50 = 0.82 ± 0.18 nM) were ~9-fold and ~2-fold more potent than U50,488H (EC50 = 1.61 ± 0.19 nM), respectively. For β-arrestin recruitment, the maximal effect of nalfurafine (Emax = 108.0 ± 2.6%) was higher than that of U50,488H (Emax = 99.93 ± 0.07%), and the maximal effect of 42B (Emax = 116.8 ± 1.0%) was higher than those of nalfurafine and U50,488H. While the potency of nalfurafine (EC50 = 0.74 ± 0.07 nM) was not significantly different from that of U50,488 (EC50 = 1.43 ± 0.13 nM), 42B (EC50 = 3.22 ± 0.42 nM) showed ~2-fold and ~4-fold lower potency for β-arrestin recruitment than U50,488H and nalfurafine, respectively.
Figure 3. KOR agonists-induced G protein activation (GloSensor cAMP assay) and β-arrestin recruitment (Tango assay).
GloSensor cAMP and Tango assays were performed by PDSP of the NIMH, which is directed by Dr. Bryan Roth of University of North Carolina (Web site: https://pdsp.unc.edu/ims/investigator/web/). EC50 values and maximal responses are shown in Table 2. Each value is mean ± SEM (n=3).
Table 2.
Summary of EC50 and Emax values of KOR agonists for G protein activation as measured by GloSensor cAMP assay and G protein independent β-arrestin recruitment as measured by Tango assay. The prototypic selective KOR agonist U50,488H was used as the balanced agonist. Emax values in the GloSensor assay denote the maximal degree of inhibition. Data were derived from the curves in Figure 3. Mean ± SEM (n = 3). EC50 and Emax data of GloSensor cAMP and Tango assays were analyzed with one-way ANOVA followed by Tukey’s post-hoc test, respectively. Results of one-way ANOVA are: Emax of GloSensor cAMP assay: F (2,6) = 2.77, p > 0.05; EC50 of GloSensor cAMP assay: F (2,6) = 22.91, p < 0.01; Emax of Tango assay: F (2,6) = 28.32, p < 0.001;
GloSensor cAMP assay | Tango assay | |||||
---|---|---|---|---|---|---|
EC50(nM) | Emax(%) | N | EC50(nM) | Emax | N | |
U50,488H | 1.61±0.19 | 72.95±0.86 | 3 | 1.43±0.13 | 99.93±0.07 | 3 |
Nalfurafine | 0.17±0.04** | 69.82±1.53 | 3 | 0.74±0.07 | 108.0±2.6* | 3 |
42B | 0.82±0.18* | 74.40±1.68 | 3 | 3.22±0.42**,## | 116.8±1.0***,# | 3 |
EC50 of Tango assay: F (2,6) = 24.80, p < 0.01. Significance levels are
p < 0.05,
p < 0.01,
p < 0.001, compared with U50,488H in the same assay;
p < 0.05,
p < 0.01, compared with nalfurafine in the same assay, by Tukey’s post-hoc test.
The intrinsic relative activity (RAi) of the agonists for G protein activation (RAi−G) and β-arrestin recruitment (RAi−b) were calculated from the agonist concentration-response curves (Figure 3) and summarized in Table 3, using the method of Ehlert and colleagues25–27. This parameter represents the product of affinity (1/Kobs) and efficacy (ε) of a test agonist, divided by the corresponding product for a reference agonist (ε’/Kobs’) (RAi = εKobs’/ε’Kobs). In our experiments, we designated U50,488H as the reference agonist. RAi was estimated using global nonlinear regression analysis with a modified form of the operational model26, 27. It has been shown that RAi is a relative estimate of active state affinity, which is why RAi is useful in quantifying biased signaling. The bias factor represents the ratio of the RAi value for G protein signaling divided by that for arrestin signaling, and these values are also summarized in Table 3. In the present study, the agonists exhibit Hill slopes <1 in the GloSensor assay (0.64 – 0.74) and Hill slopes >1 in the Tango assay (1.1, 1.4), which were taken into consideration in our calculations.
Table 3.
log RAi−G, log RAi−b, log (RAi−G/RAi−b), bias factors to G protein signaling and probability values of KOR agonists. The parameters were calculated from the same agonist concentration-response curves that were used to calculate EC50 and Emax values (Figure 3, Table 2) using the method described by Ehlert and colleagues25–27. The prototypic selective KOR agonist U50,488H was designated as the “balanced” or standard reference ligand. The bias factor to G protein signaling for a given ligand is defined as the ratio of RAi−G divided by RAi−b. Mean ± SEM (n = 3).
Log RAi-G | Log RAi-b | G Biased factor (95% CI) | (p value)* | ||
---|---|---|---|---|---|
U50,488H | 0 | 0 | 0 | 1 | |
Nalfurafine | 0.94±0.14 | 0.28±0.02 | 0.65±0.40 | 4.49 (1.79–11.1) | 0.010 |
42B | 0.17±0.06 | −0.28±0.03 | 0.46±0.20 | 2.85 (1.82–4.48) | 0.003 |
G, G proteins; b, β-arrestin; CI: confidence interval.
compared to bias factor of 1 by t test.
A bias factor > 1 indicates a preference for G protein signaling, whereas a bias factor < 1 indicate a preference for β-arrestin recruitment. As shown in Table 3, the bias factors of nalfurafine and 42B were 4.49 and 2.85, respectively, suggesting that both nalfurafine and 42B have small, significant biases for G protein signaling.
Thus, these results indicate that both nalfurafine and 42B are biased agonists for G protein signaling. Earlier research on whether nalfurafine is a biased agonist yielded different results, however, including G protein-biased12, 28, un-biased11 and β-arrestin-biased29. We previously demonstrated that in vitro nalfurafine was not biased using [35S]GTPγS binding and β-arrestin1 and β-arrestin2 recruitment as the end points11. Thus, there are discrepancies between our own two studies. In these earlier studies, for [35S]GTPγS binding we used mouse neuro2A cells stably transfected with the mouse KOR and for β-arrestin recruitment assay we used β-galactosidase fragment complementation assay and HEK293 cells stably transfected with the mouse KOR conjugated with β-galactosidase donor fragment and β-arrestin-1 or β-arrestin-2 fused with β-galactosidase acceptor fragment11. In the present study, experiments were performed by the PDSP program, in which HEK293-T cells stably expressing the human KOR were transfected with the GloSensor cAMP DNA construct for GloSensor cAMP assay and HEK293 cells stably expressing a tTA-dependent luciferase reporter and a β-arrestin2-TEV fusion gene were transfected with the human KOR for Tango assays. In both studies, U50,488H was used as the reference balanced agonist. Thus, the assay, cell line, species origin of the receptor, receptor expression level and kinetics of agonist-receptor interaction may contribute to the differences, as discussed previously30–33. The recent finding of Gillis, et al.34, which revealed that responses that have amplification factors and have higher receptor reserves tend to enhance potency and efficacy of an agonist, thus confounding agonist bias calculation. The inhibition of adenylyl cyclase response has a high signal amplification34, whereas [35S]GTPγS binding and β-arrestin recruitment do not, thus making nalfurafine to be G protein-biased in the present study, not in our previous one11. In addition, while both Schattauer et al.28 and Kaski et al.12 reported nalfurafine to be a G protein-biased agonist using various assays for G protein and β-arrestin signaling in human or rat KOR-expressing HEK293 or HEK293T cells with U50,488 as the reference balanced agonist, Dunn, et al.29 showed that nalfurafine was a β-arrestin-biased agonist using [35S]GTPγS binding and β-arrestin2 recruitment assays in human KOR-expressing U2OS cells with U69,593 as the reference balanced agonist. Again, the inconsistency may be attributed to different assays, cell lines and the reference agonist used and the kinetics of agonist action can be a factor if not addressed by the experimental protocol. In addition to amplification factors of signaling end points, kinetics of agonist action on the receptor also is a factor, thus end points of assays also play important roles in determining the bias factors.
Molecular docking studies
Nalfurafine and 42B have identical chemical structure skeletons, except for 3-hydroxy group on the phenyl moiety of nalfurafine (Figure 1). Nalfurafine and 42B were docked into the agonist-bound crystal structures of the MOR (PDB ID: 5C1M)35 and KOR (PDB ID: 6B73)24 to explore how nalfurafine and 42B may interact with the receptors so that they produce different potency profiles for the MOR and KOR. The docking poses with the highest CHEM-PLP scores were selected as the optimal binding poses, named as nalfurafine_MORactive, nalfurafine_KORactive, 42B_MORactive, and 42B_KORactive complexes (Figure 4 a–d), respectively.
Figure 4. Binding poses of nalfurafine and 42B with the active MOR and KOR from molecular docking studies.
(a) nalfurafine_MORactive, (b) nalfurafine_KORactive, (c) 42B_MORactive, (d) 42B_KORactive. The MOR and KOR shown as cartoon models in light-blue and light-pink, respectively. Nalfurafine, 42B, and key amino acid residues shown as stick models. Carbon atoms: nalfurafine in green; 42B in cyan; key amino acid residues of the MOR in yellow, KOR in gray. The red dashed line represented possible hydrogen bonds. The water molecules shown as sphere models in magenta.
From the docking study results, nalfurafine and 42B in the active MOR and KOR adopted similar binding modes as BU72 and MP1104 did in the crystal structures of the active MOR (PDB ID: 5C1M)35 and KOR (PDB ID: 6B73)24, respectively (Supporting Information, Figure S1 a–b). Especially, the cyclopropylmethyl moieties of nalfurafine and 42B were accommodated by the hydrophobic pockets formed by residues W6.48, G7.42, and Y7.43 of the MOR and KOR. The furan moieties of nalfurafine and 42B seemed to interact with the transmembrane helices 2 and 3 (TMs 2 and 3). Notably, these interactions were thought to stabilize the active states of the MOR and KOR (Figure S1 a–b)24, 35. Therefore, that may help explain why nalfurafine and 42B exhibited agonist activity to the MOR and KOR.
Further zooming in, there were at least 22 residues within 5 Å of nalfurafine and 42B in the nalfurafine_MORactive, nalfurafine_KORactive, 42B_MORactive, and 42B_KORactive complexes. As shown in Figure 4, conserved residues M3.36, W6.48, I6.51, H 6.52, and I7.39 located at the orthosteric binding sites may form direct hydrophobic interactions with the epoxymorphinan scaffold of both ligands. In addition, the highly conserved D3.32 may form ionic interactions with the 17-quaternary nitrogen atom of nalfurafine and 42B, and Y3.33 may form hydrogen bond interactions with the dihydrofuran oxygen atom of both ligands (Table S2). Notably, among the 22 residues, two residues (K6.58 and W7.35 in the MOR, and E6.58 and Y7.35 in the KOR) were non-conserved ones. In the nalfurafine_KORactive and 42B_KORactive complexes, Y7.35 may form hydrogen bonds with the carbonyl groups of nalfurafine and 42B (Table S2), which may help stabilize the binding of the furan moieties of nalfurafine and 42B. However, this hydrogen bond was not observed with W7.35 in the nalfurafine_MORactive and 42B_MORactive complexes. As discussed above, the furan moieties of nalfurafine and 42B interacting with the TMs 2 and 3 was involved in the activation of the MOR and KOR. The hydrogen bonds formed between Y7.35 in the KOR and the carbonyl groups of nalfurafine and 42B may provide a possible explanation for the observations that nalfurafine and 42B exhibited higher binding affinities with the KOR than the MOR while both acted as KOR full agonists, but MOR partial agonists.
In addition, as described in the crystal structures of the active MOR (PDB ID: 5C1M)35 and KOR (PDB ID: 6B73)24, water-mediated hydrogen bonding interactions were formed between the hydroxy groups of the phenolic moiety of the ligands, BU72 and MP1104, and conserved residues Y3.33, K5.39, and H6.52, respectively (Figure S1). These interactions may retain the position of the side chain of W6.48 and further stabilize the active state of the MOR and KOR. Moreover, in the nalfurafine_KORactive complex, the same water-mediated hydrogen bonding interactions were observed between the 3-hydroxy group of the phenolic moiety of nalfurafine and residues Y3.33, K5.39, and H6.52 (Table S2), while there were no water-mediated hydrogen bonding interactions in the 42B_KORactive complex due to the lack of the 3-hydroxy group in 42B. Therefore, the lack of the water-mediated hydrogen bonding interaction in the 42B_KORactive complexes, compared with that of the nalfurafine_KORactive complex, may be the cause of lower potency of 42B at the KOR, compared to nalfurafine.
42B produced antinociceptive and anti-scratching effects in a dose-dependent manner, similar to nalfurafine.
Activation of the KOR produces antinociceptive and anti-scratching effects1, 3, 11. We examined activities of 42B in inhibiting pain-like behaviors in the formalin test11, 36 and in attenuating compound 48/80-induced scratching3, 11, 17.
42B (1, 3, 5 mg/kg) reduced licking time in phase II of the formalin test in a dose-dependent manner in mice, indicating antinociceptive effects (Figure 5a). The A50 value was determined to be 2.08 mg/kg. Our previous results demonstrated that nalfurafine dose-dependently reduced pain behavior in the late phase of the formalin test (Figure 5b), with an A50 value of 5.8 μg/kg11.
Figure 5. 42B produced antinociceptive and anti-scratching effects in mice, like nalfurafine.
(a) and (b) 42B inhibited formalin-induced pain behaviors in mice like nalfurafine. Saline or one of several doses of 42B or nalfurafine was injected (s.c.) 5 min before formalin and the amount of time the animal spent licking the injected paw was counted for 20 min starting at 15 min after formalin injection. A50 doses were determined as described11. Data were analyzed using one-way ANOVA followed by Dunnett’s post-hoc test. Results of one-way ANOVA are: 42B, F(3,24) = 41.63, p < 0.001; nalfurafine, F(4, 41) = 18.65, p < 0.0001. Significance levels are *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001, compared to saline control, by Dunnett’s post-hoc test (mean ± SEM, n = 6–10 animals/group). Data on nalfurafine were from Liu, et al.11 and are shown for comparison.
(c) and (d) 42B induced inhibition of scratching behavior induced by compound 48/80, like nalfurafine. Saline or one of the different doses of 42B or nalfurafine was injected (s.c.) 20 min before compound 48/80 and the bouts of scratching were counted for 30 min. A50 doses were determined as described previously11. Data were analyzed using one-way ANOVA followed by Dunnett’s post-hoc test. Results of one-way ANOVA were: 42B, F(3,28) = 11.68, p < 0.001; nalfurafine, F(5,44) = 31.04, p < 0.0001. Significance levels are **p < 0.01, ***p < 0.001, ****p < 0.0001, compared to saline control by Dunnett’s post-hoc test (mean ± SEM, n = 6–12 animals/group). Data on nalfurafine were from Liu, et al.11 and are shown for comparison.
(e) The selective KOR antagonist norBNI blocked anti-scratching effects of 42B in compound 48/80 scratching test. Saline or norBNI was injected (20 mg/kg, i.p.) 20 h before saline or 42B, 20 min later compound 48/80 was injected and the bouts of scratching were counted for 30 min. Data were analyzed with two-way ANOVA followed by Sidak’s post-hoc test. Results of two-way ANOVA showed a significant main effects of 42B [F(1,21) = 13.26, p < 0.01], a significant main effects of norBNI [F(1,21) = 7.47, p < 0.05] and a significant interaction [F (1,21) = 5.39, p < 0.05]. Significance levels are **p < 0.01, compared to saline+saline group; ##p < 0.01, compared to saline+42B group, by Sidak’s post-hoc test (mean ± SEM, n = 5–8 animals/group).
42B (1, 3, 5 mg/kg) significantly inhibited compound 48/80-induced scratching in a dose-dependent manner in mice, indicating anti-scratch effects. The A50 value was determined to be 2.82 mg/kg. Similarly, we previously showed that nalfurafine (2.5, 5, 10, 20, 30 μg/kg) displayed dose-dependent anti-scratch effect in mice with an A50 value of 8.0 μg/kg11 (Figure 5c,d).
Thus, nalfurafine was 400x and about 340x more potent than 42B, respectively, in the formalin test and in the anti-scratch test, consistent with the differences in their in vitro potency (260X) for the KOR in [35S]GTPγS binding. However, in inhibition of forskolin-stimulated adenylyl cyclase (Glosensor assay), nalfurafine was only ~5X more potent than 42B.
We then examined if 42B produced anti-scratching effects via KOR activation by pretreatment of mice with the selective KOR antagonist norBNI (20 mg/kg, i.p.) 20 h prior to saline or 42B injection. While norBNI alone had no effects on compound 48/80-induced scratching, administration of norBNI 20 h before 42B (3 mg/kg) eliminated the anti-scratch activity of 42B, indicating that 42B produces its anti-scratching effects through activation of the KOR (Figure 5e).
Effects of 42B on adverse behaviors induced by KOR activation
Motor incoordination and sedation
KOR activation results in impaired performance in the rotarod test and inhibition of novelty-induced hyperlocomotion11, 37, measures of motor incoordination and sedation in rodents. We tested if 42B would produce these effects in mice.
42B (1, 3, 5 mg/kg) dose-dependently caused performance impairment in the rotarod test (Figure 6a), indicating motor incoordination. Even at 1 mg/kg, which is lower than its A50 values in the antinociceptive and anti-scratch tests, 42B induced significant effect on rotarod test at 10 min. Interestingly, the effect of 42B was short-lived. The peak effect was at 10 min among the time points tested, and returned to the control level at 40 min. We previously showed nalfurafine at 20 μg/kg nalfurafine had small, yet significant, effects at 30 min and 40 min following drug injection11. The prototypic KOR agonist U50,488H was used as a positive control. The effect induced by 20 μg/kg is like that of U50,488H at 2 mg/kg, but much attenuated than 5 mg/kg of U50,488H (Figure 6b). U50,488H (5 mg/kg) significantly reduced the time mice stayed on the rod at 10, 20, 30, and 40 min after drug administration and the effect remained robust at 40 min. The doses of 20 μg/kg of nalfurafine and 5 mg/kg of U50,488 were 2.5 × their A50 values in the anti-scratching test11.
Figure 6. Comparison of 42B and nalfurafine in KOR agonist-induced adverse behaviors: motor incoordination, sedation, and conditioned place aversion (CPA) in mice.
(a) and (b) unlike nalfurafine, 42B caused motor incoordination in the rotarod test in mice. After training the previous day, mice were injected s.c. with saline, U50,488H (2, 5 mg/kg), 42B (1, 3, 5 mg/kg) or nalfurafine (20 μg/kg) and tested on the rotarods 10, 20, 30, and 40 min after injection. The time each stayed on the rods was recorded and normalized against the baseline. Data were analyzed with two-way ANOVA followed by Dunnett’s post-hoc test (mean ± SEM, n= 9–12/group). 42B: Results of two-way ANOVA showed a significant main effect of treatment [F (3,36) = 10.48, p < 0.0001], a significant main effect of time [F (4,144) = 32.33, p < 0.0001] and a significant interaction [F (12,199) = 4.65, p < 0.0001]. Nalfurafine: Results of two-way ANOVA showed a significant main effect of treatment [F (3,36) = 15.43, p < 0.0001], a significant main effect of time [F (4,144) = 24.14, p < 0.0001] and a significant interaction [F (12,199) = 5.46, p < 0.0001]. *p < 0.05, **p < 0.01, ****p < 0.0001, compared with 0 min of each group; #p < 0.05, ##p < 0.01, ####p < 0.0001, compared with the saline group at the same time, by Dunnett’s post-hoc test. Data on nalfurafine and 5 mg/kg U50,488H were from Liu, et al.11 and are shown for comparison.
(c) and (d) Unlike nalfurafine, 42B caused inhibition of novelty-induced locomotor activity in mice. Mice were treated s.c. with saline, 42B (1, 3, 5 mg/kg), U50,488H (5 mg/kg), or nalfurafine (20 μg/kg) (2.5x A50 values in the anti-scratching test) and locomotor activities were monitored. Cumulative data between 0–30 min post-injection are shown here. Each value represents mean ± SEM (n = 8–14). (c) Ambulatory activity of 42B: ***p < 0.001, compared with the saline group by one-way ANOVA [F (3,34) = 7.59, p < 0.001] followed by Dunnett’s post-hoc test. (d) Ambulatory activity of U50,488H and nalfurafine: *p < 0.05, compared with the saline group by one-way ANOVA [F (2,27) = 6.254, p < 0.01] followed by Dunnett’s post-hoc test. Data on nalfurafine and 5 mg/kg U50,488H were from Liu, et al.11 and are shown for comparison.
(e) and (f) 42B did not cause CPA in mice, like nalfurafine. On Day 0, mice were subject to pre-test. On Days 1–6, Mice were injected with saline or one of the various doses of U50,488H, 42B or nalfurafine before each 30-min conditioning session (1 saline session and 1 drug session/day) for 6 days. On Day 7 (post-test), the length of time the animal spent on the drug-paired side was measured. The graph shows the time the animal spent during the post-test subtracting the amount of time spent during the pre-test. Data were analyzed with one-way ANOVA followed by Dunnett’s post-hoc test (for the 42B and nalfurafine experiments) and unpaired t-test (for U50,488H) (mean ± SEM, n = 9–10/group). 42B, F(3,36) = 0.45, p > 0.05; nalfurafine, F(4,47) = 0.38, p > 0.05. Significance levels are **p < 0.01, compared to saline control by unpaired t-test. Data on nalfurafine and U50,488H were from Liu, et al.11 and are shown for comparison.
(g) Naloxone at a dose (1 mg/kg) selective for the MOR did not alter lack of CPA of 42B. On Day 0, mice were subject to pre-test. On Days 1–6, saline or naloxone was injected 25 min before saline or 42B, then 30-min conditioning session began immediately (1 saline session and 1 drug session/day) for 6 days. U50,488H alone was used as the CPA positive control. On Day 7 (post-test), the length of time the animal spent on the drug-paired side was measured. The graph shows the time the animal spent during the post-test subtracting the amount of time spent during the pre-test. Data were analyzed with two-way ANOVA followed by Sidak’s post-hoc test (for the 42B-naloxone experiments) and unpaired t-test (for U50,488H) (mean ± SEM, n = 10/group). Results of two-way ANOVA showed no significant main effects of 42B [F(1,36) = 0.13, p > 0.05], naloxone [F(1,36) = 0.004, p > 0.05] or interaction [F (1,36) = 1.16, p > 0.05]. Significance levels are **p < 0.01, compared to saline + saline by unpaired t-test.
42B (1, 3, 5 mg/kg) caused significant and dose-dependent reduction in novelty-induced hyperlocomotion in mice (Figure 6c). U50,488 at 5 mg/kg showed significant inhibition, whereas nalfurafine at 20 μg/kg did not (Figure 6d)11.
Thus, unlike nalfurafine, 42B caused motor incoordination and sedation in the dose range producing antinociceptive and anti-scratching effects.
CPA
KOR agonists have been shown to cause dysphoria and psychotomimetic effects in humans2, 4, 5 and CPA in rodents11, 38, 39. We examined if 42B caused CPA. 42B did not cause CPA in mice at the doses (1, 3, 5 mg/kg) producing antinociceptive and anti-scratching effects (Figure 6e). We reported previously that nalfurafine did not produce CPA even at 20 μg/kg, while U50,488 (5 mg/kg) induced significant CPA, compared to the saline group (Figure 6f)11. The doses of nalfurafine and U50,488H are 2.5-fold of the A50 in the anti-scratch test11. Thus, like nalfurafine, 42B did not produce CPA test in mice.
42B showed only 8.4-fold functional selectivity for the KOR over the MOR in vitro. Lack of CPA by 42B may be due to a combination of its actions on both the MOR and the KOR, causing conditioned place preference (CPP) and CPA, respectively. We tested this possibility by blocking the MOR with naloxone at a dose (1 mg/kg) which is selective for the MOR. Neither naloxone (1 mg/kg) alone nor a combination of naloxone (1 mg/kg) and 42B (3 mg/kg) caused CPA or CPP, compared to the saline group, suggesting that MOR did not contribute to lack of CPA by 42B (Figure 6g). As a positive control in this experiment, U50,488 (5 mg/kg) induced significant CPA.
Our results that neither nalfurafine up to 20 μg/kg nor 42B up to 5 mg/kg produced CPA demonstrated that there is dose separation between antinociception/anti-scratch and CPA for both nalfurafine11, 18, 40 and 42B.
42B acts on the KOR, but not the MOR, to produce its behavioral effects
42B showed relatively low KOR/MOR selectivity. The following evidence strongly suggests that 42B likely acts on the KOR to produce its in vivo effects. The anti-scratch effect of 42B was blocked by norBNI pretreatment (20 mg/kg, i.p.) for 20 h or longer (see Figure 5e). Such a norBNI treatment paradigm was shown to selectively block the KOR, but not MOR or DOR41–43, indicating that 42B at 3 mg/kg (and likely all the doses we used) acts on the KOR. In addition, KOR agonists and MOR agonists produce different effects in some behavior end points in mice. KOR agonists, but not MOR agonists, produce anti-scratch effect17, 21, 44. Moreover, in mice KOR agonists produce hypolocomotion, whereas MOR agonists cause hyperlocomotion11, 37, 45. Here we demonstrated that 42B induced anti-scratch effect and hypolocomotion, indicating KOR-mediated effects. In addition, blockade of MOR by naloxone (1 mg/kg, s.c.) did not affect its lack of CPA by 42B (see Figure 6g). We did not use the selective MOR antagonist CTAP or CTOP because these compounds required intracerebroventricular injection. Naloxone has a higher affinity for the MOR than for the KOR. In vivo, naloxone is 20–36 times more potent in antagonizing morphine- than U50,488- or spiradoline-induced antinociception in the mouse tail flick test1, 46. A low dose (0.5 – 1 mg/kg) of naloxone has been often used to determine the role of MOR in physiological or pharmacological responses (for example,47–49).
Nalfurafine and 42B showed similar G protein biases, but had different effects on locomotor activity and motor coordination
It is believed that G protein- and β-arrestin-mediated signaling pathways were linked to different KOR-mediated behaviors. Deletion of β-arrestin2 in mice impaired KOR-mediated motor incoordination in the rotarod test, but did not affect antinociceptive effects in the hot plate test, CPA and hypolocomotion produced by the KOR agonists U69,593 or salvinorin A37. Neither did it affect inhibitory effect of U50,488H on chloroquine-induced scratching50. These results indicate that β-arrestin2 is involved in motor incoordination in the rotarod test, but has no role in KOR-mediated analgesic effect (in the hot plate test), anti-scratch effect, CPA and hypolocomotion, and suggest that these effects may be mediated by G protein signaling. However, the possibility cannot be excluded that β-arrestin1 may compensate for the absence of β-arrestin2 in these mice. In contrast, KOR activation produces GRK3-dependent p38 MAPK activation51 and inhibition of this pathway blocked KOR-mediated CPA52–54, suggesting that CPA is mediated by GRK3- and arrestin-dependent pathway.
In this study, we demonstrated that both nalfurafine and 42B had biases towards G protein signaling in vitro and did not cause CPA, which is different from the findings of White et al. that G protein-biased RB64 caused CPA. However, 42B, unlike nalfurafine, produced motor incoordination and hypolocomotion, which is opposite to the previous finding that β-arrestin2 is involved in motor incoordination. Furthermore, Dunn, et al.29 found that impaired rotarod performance is correlated with β-arrestin bias by examining nine KOR agonists of different biases in the rotarod test29. These inconsistent observations underscore an important consideration, that is, one should be cautious in correlating in vitro biochemical data with in vivo behavior results. As discussed above, different in vitro assay conditions yielded varying bias results for the same compound.
In addition, prior studies of distinct G protein-biased KOR agonists showed varying pharmacological properties, further indicating that bias factors in vitro may not predict pharmacological properties of agonists in vivo. Bohn and colleagues reported that the selective KOR agonists triazole 1.1 and Isoquinolinone 2.1 showed G protein biases, using U69,593 or U50,488H as the reference balanced agonist55, 56. These two G protein-biased KOR agonists produced antinociception in the warm water tail withdrawal test and inhibited chloroquine-promoted scratching similar to U50,488H, but they did not decrease locomotor activity in the open field test50, 55, 56. Roth and colleagues reported that RB-64, a salvinorin A derivative, is a G protein-biased KOR agonist, with salvinorin A as the reference balanced agonist37. RB-64 produced antinociceptive effect in the hot plate test, caused CPA, but no motor incoordination in the rotarod test and no hypolocomotion37. In addition, triazole 1.1 and RB-64 did not cause an increase in baseline threshold in the intracranial self-stimulation test as U50,488H did37, 56, suggesting lack of anhedonia; however, U50,488H, as a positive control in the ICSS test, decreased the maximal spinning rate, confounding data interpretation. Taken together, among the five KOR agonists reported to have G protein-biases (triazole 1.1, Isoquinolinone 2.1, RB-64, nalfurafine and 42B), all produced antinociception, four had anti-scratch effects (RB-64 not examined) and four did not affect locomotor activity (42B did). In the rotarod test, RB-64 had no effect, nalfurafine had a slight effect, but 42B showed profound effect (triazole 1.1 and isoquinone 2.1 not tested). While RB-64 caused CPA, nalfurafine and 42B did not.
There are many other factors involved in in vivo pharmacological effects of drugs besides in vitro pharmacological properties. Pharmacokinetic characteristics of a drug are critical in determining its in vivo actions. Our bias estimates are made in vitro to circumvent pharmacokinetic issues. The log RAi estimate is a relative estimate of active state affinity, which is why the parameter is used to estimate agonist-receptor bias for different signaling pathways. Even at the pharmacodynamic level, the in vitro bias estimate does not directly translate to in vivo behavior of agonists for three reasons, 1) it is a relative estimate and the reference agonist may not be strictly neutral for different pathways, 2) the sensitivities of the signaling pathways measured in vivo may be different from those measured in vitro (sensitivity refers to how much redundancy and amplification occurs in the pathway), 3) the signaling pathway measured in vitro may be different from that measured in vivo. In addition, pharmacokinetic issues may alter the potencies of agonists in vivo, such as absorption, distribution, metabolism into active or inactive metabolites and excretion following drug administration. In the case of drugs acting on the CNS, the degree of drugs crossing blood-brain barrier is also an important consideration. 42B appears to get into the brain because it caused hypolocomotion and motor incoordination. Nalfurafine was shown to get into the brain previously. Systemic nalfurafine suppressed intracisternal or intrathecal morphine-induced scratching21, 57 and affected several signaling pathways in brain regions11. The anti-scratch activity of systemic nalfurafine was blocked by intra-cerebroventricularly injected norBNI21. It is conceivable that there are differences between the two drugs in some pharmacokinetic parameters, such as membrane partitioning, tissue binding, time course of the free drug concentrations in brain and volume of distribution. Pharmacokinetic properties of 42B have not been characterized, which is outside of the scope of our current study. Nalfurafine was shown to be a P-glycoprotein substrate58, decreasing its influx into the brain. 3-OH group is likely to play a role in P-glycoprotein efflux, thus 42B may have higher influx into the brain and likely have higher brain concentration. In the rotarod test, 42B produced profound inhibitory effect with the peak effect at 10 min, whereas nalfurafine caused its slight lowering effect with a peak at 30 min, suggesting that 42B is distributed into the brain more readily than nalfurafine. Nalfurafine was shown to be metabolized by N17-decyclopropylmethylation and 3-OH glucuronidation59. 42B, lacking the 3-OH group, is more lipophilic and also will not undergo glucuronidation and may impact on its half-life in the blood. Thus, pharmacokinetic properties of the drug, particularly the time course of the free drug concentrations in brain, play critical roles in its pharmacological effects in vivo. Most studies on in vivo effects of biased agonists defined in vitro did not examine detailed pharmacokinetic properties of the compounds.
The behavioral effects are likely driven by the brain concentration of the drug and therefore temporally sensitive. As mentioned above, the effects of 42B and nalfurafine in the rotarod test reached peaks at 10 min and 30 min after injection, respectively, which likely approximated the times of the maximal concentrations of the drugs. The endpoints we examined were measured cumulatively for the following time periods post drug injection: from 20 to 40 min for the formalin test, from 20 to 50 min for the anti-scratch test, from 0 to 30 min for the locomotor activity, from 0 to 40 min for the rotarod test and from 0 to 30 min for the conditioning phase of the CPA test (similar results were obtained when conditioning was done from 10 to 40 min). Thus, we may underestimate the potency/efficacy or 42B in the formalin test and the anti-scratch test, but we have captured the peak effects of both nalfurafine and 42B in all the other tests.
Our observations that 42B produced hypolocomotion and motor incoordination, but not CPA, suggest that the signaling mechanism underlying CPA is different from those of hypolocomotion and motor incoordination. These results are in line with our previous observation that rapamycin, a mTOR inhibitor, abrogated U50,488H-promoted CPA, but not hypolocomotion and motor incoordination, in mice11. Our findings demonstrated more complexity in KOR-mediated behaviors which can be grouped based on mechanisms involved: analgesia / anti-scratch effect; hypolocomotion / motor incoordination; CPA. The separation of motor incoordination from CPA is consistent with the findings of White, et al.37.
Conclusion
Our in vitro data that 42B had lower KOR/MOR selectivity than nalfurafine are different from published data. The observations that 42B showed lower affinity for the KOR and MOR and lower KOR/MOR selectivity were explained by molecular modeling results on their interactions with active states of the MOR and KOR, highlighting the importance of 3-OH group of nalfurafine. Furthermore, both nalfurafine and 42B showed biases towards G protein signaling. Like nalfurafine, 42B produced analgesic and anti-scratching effects, and did not cause CPA in mice. In addition, antipruritic effects of 42B was KOR-mediated and MOR did not contribute to lack of CPA by 42B, like nalfurafine. In contrast, unlike nalfurafine, 42B impaired motor coordination and induced sedation in mice. Therefore, 3-hydroxy group of nalfurafine is important for its unique and favorable pharmacological profile. Among the KOR-induced adverse behaviors, CPA was separated from motor incoordination and hypolocomotion. As both agonists showed G protein biases yet produced different effects on locomotor activity and motor incoordination, the findings and those in the literature suggest caution in correlating in vitro biochemical data with in vivo behavior effects.
Materials and Methods
Drugs and Materials
(−)U50,488 (U111), DAMGO (E-7384), DPDPE (E3888) and Nociceptin / OFQ (487960) were purchased from Sigma-Aldrich (St. Louis, MO). Nalfurafine was obtained from the National Institute on Drug Abuse (Bethesda, MD). 42B was synthesized by the laboratory of Dr. Yan Zhang of Virginia Commonwealth University (Richmond, VA) (See Supporting Information for details in chemical synthesis). [35S]GTPγS (1250 Ci/mmol) was purchased from PerkinElmer Life Sciences (Boston, MA). The following materials were purchase from Sigma-Aldrich (St. Louis, MO): formalin, compound 48/80, GDP and GTPγS. Other commonly used chemicals were obtained from Sigma-Aldrich or ThermoFisher Scientific.
Cell Lines
CHO cells that stably express the rat MOR60, the mouse DOR61, the human KOR62 or the human NOR were cultured in Minimum Essential medium (Gibco, Grand Island, NY) supplemented with 10% fetal bovine serum (Gibco), 200 μg/ml geneticin, 100 U/ml penicillin and 100 U/ml streptomycin (Sigma-Aldrich, St. Louis, MO) at 37 °C in a humidified atmosphere containing 5% CO2. CHO cells stably expressing the mouse DOR were kindly supplied by Dr. Ping-Yi Law, formerly Department of Pharmacology, University of Minnesota School of Medicine, Minneapolis, MN. CHO cells stably transfected with the human NOR were a gift from Dr. Lawrence Toll, formerly SRI International, Menlo Park, CA and currently Florida Atlantic University63. Others were established as we described previously64, 65.
Cell Membrane Preparation and [35S]GTPγS Binding
Membranes were prepared according to a modified procedure of Huang, et al.66. Cells were washed twice and harvested in Versene solution (EDTA 0.54 mM, NaCl 140 mM, KCl 2.7 mM, Na2HPO4 8.1 mM, KH2PO4 1.46 mM, and glucose 1 mM) and centrifuged at 130 × g for 5 min. The cell pellet was suspended in buffer A [50 mM Tris (pH 7.4), 1 mM EDTA, and 0.1 mM phenylmethylsulfonyl fluoride] and kept on ice for 30 min, then centrifuged at 50,000 × g for 30 min. The pellet was re-suspended in buffer A and centrifuged again. Then, the membrane pellet was re-suspended in buffer A, homogenized and then aliquoted at about 1.5 mg/ml, frozen in dry ice/ethanol, and stored at −80℃. All the procedures were performed at 4°C.
Determination of [35S]GTPγS binding to G proteins was carried out using a modified procedure of Huang, et al.66. Immediately before the [35S]GTPγS binding assay, membranes were thawed on ice, sonicated and diluted with buffer B [50 mM Tris (pH 7.4), 100 mM (KOR) or 50 mM (MOR, DOR or NOR) NaCl, 5 mM MgCl2, and 1 mM EDTA]. Membranes (10 μg) were incubated with [35S]GTPγS (80 pM, 190,000–220,000 dpm) and 15 μM GDP with or without a ligand (10−13 to 5 × 10−5 M) in a total volume of 1 ml buffer B for 60 min at 30℃. Nonspecific binding was defined by incubation in the presence of 10 μM GTPγS. Bound and free [35S]GTPγS were separated by filtration with GF/B filters under reduced pressure. Radioactivity on filters was determined by liquid scintillation counting. EC50 values and Emax of drugs were determined by curve fitting to the equation for a sigmoidal curve using GraphPad Prism version 8.2.1 (GraphPad Software Inc., La Jolla, CA).
Molecular docking studies
As nalfurafine and 42B (Figure 1a,b) are MOR partial agonists and KOR full agonists17, the crystal structures of active MOR (PDB ID: 5C1M)35 and KOR (PDB ID: 6B73)24 were downloaded from the Protein Data Bank at http://www.rcsb.org to be applied as target protein templates. Before conducting the molecular docking studies, the N terminus, the seven transmembrane helices, the C terminus, and the three crystal water molecules involved in the water-mediated interaction between residues Y3.33, K5.39, and H6.52 and the hydroxyl group of the original ligands, BU72 and MP1104, were retained24, 35, 67–70, and other molecules were removed from both crystal structures. The missing residues in extracellular loop 2 and intracellular loop 3 of 6B73 were then constructed and implemented through Sybyl 8.071. The missing hydrogen atoms were added to both receptors. Meanwhile, nalfurafine and 42B were sketched, assigned with Gasteiger-Hückel charges, and further energy minimized to a gradient of 0.05 with 10,000 iterations in Sybyl 8.071.
The molecular docking studies were conducted by using GOLD 5.6. Similar to the studies of the epoxymorphinan analogs developed in our group24,35, 72–77, D3.32 in the MOR and KOR may form ionic interactions with the protonated nitrogen atom at the 17-amino group of nalfurafine and 42B. Y3.33 in the MOR and KOR may form hydrogen bonding interactions with the dihydrofuran oxygen atom of both ligands. Thus, the binding sites of the MOR and KOR were defined by the atoms within 10 Å of the γ-carbon atom of D3.32 in the MOR and KOR, respectively. The distances between the protonated nitrogen atom in the 17-amino group of nalfurafine and 42B and the oxygen atom at the side chain of D3.32 in the MOR and KOR were restricted to 4.0 Å. The distances between the two ligands’ dihydrofuran oxygen atom and the phenolic oxygen atom of Y3.33 were restricted to 3.5 Å. In addition, the Allow early termination option in GOLD 56 was activated during our docking studies, the number of docking solutions were set to 50. Except for above parameters, other parameters were applied their standard default settings. In the process of the molecular docking, flexible ligand was docked into a rigid protein. After the molecular docking, the docking poses with the highest CHEM-PLP score were chosen as the optimal binding poses.
Animals
Adult male CD-1 mice (30–35 g) were purchased from Charles River Laboratories (Wilmington, MA). All the mice were housed in a temperature- and humidity-controlled room on a light-dark cycle (22°C, 50–60 % humidity, 12:12 h light / dark cycle). All experiments were conducted when lights were on. The animals received food and water ad libitum, except during the experimental sessions. All procedures were approved by Temple University School of Medicine Institutional Animal Care and Use Committee.
Behavior tests
Formalin test was performed as we described11, which was based on the procedures of Murray, et al.78. Briefly, after acclimation, mice were pretreated with saline (10 ml/kg, s.c.) or 42B (1, 3, 5 mg/kg, s.c.) five minutes before 5 % formalin (20 μl, s.c.). The time each animal licked / groomed the formalin-injected paw was recorded from 15 to 35 min post-injection (phase II reaction).
Compound 48/80 scratching test was performed as we described11, which was based on the procedures of Wang, et al.17. Briefly, after acclimation, mice were injected with saline (10 ml/kg, s.c.) or 42B (1, 3, 5 mg/kg, s.c.) 20 min before compound 48/80 (50 μg, s.c), then the bouts of scratching were counted for 30 min. To determine whether the antipruritic effects of 42B is mediated by KOR, a selective KOR antagonist norBNI (20 mg/kg, i.p.) was given 20 h before saline (10 ml/kg, s.c.) or 42B (3 mg/kg, s.c.), then compound 48/80 scratching test was performed in mice.
Rotarod test was performed as we described11, which was adapted from the procedures of White, et al.37. Briefly, after one-day training, the mice with baseline > 240 s were injected with 42B (1, 3, 5 mg/kg, s.c.) or saline (10 ml/kg, s.c.) before the test, then the time each mouse stayed on the rotarod was recorded at 10, 20, 30, and 40 min after injection for a total of 5 minutes, respectively.
Measurement of locomotor activities was performed as we described11, which was according to our previous procedures79. Mice were treated with saline (10 ml/kg, s.c.) or 42B (1, 3, 5 mg/kg, s.c.) and put into locomotor chambers immediately. Activities were continuously monitored over a 60-min period.
CPA was performed as we described11, which was adapted from our procedures of CPP79. Due to the short-term effects of 42B shown in rotarod test, mice were pretreated with saline (10 ml/kg, s.c.) or 42B (1, 3, 5 mg/kg, s.c.) immediately before each conditioning session, respectively, in which mouse was confined to one chamber for 30 min. The time each mouse spent in the drug-paired chamber during pre-test and post-test was recorded, respectively. To exclude 42B’s action on MOR in lack of CPA, mice were administered with the selective MOR antagonist naloxone (1 mg/kg, s.c.) or saline (10 ml/kg, s.c.) 25 min prior to saline (10 ml/kg, s.c.) or 42B (1, 3, 5 mg/kg, s.c.), then confined to one chamber for 30 min during each conditioning session.
GloSensor cAMP and Tango assays
GloSensor cAMP and Tango assays, which are used to measure G protein activation and β-arrestin recruitment, respectively, were performed by PDSP of the NIMh, directed by Bryan Roth of University of North Carolina (Web site: https://pdsp.unc.edu/ims/investigator/web/). Briefly, HEK293-T cells stably expressing the human KOR were transfected with the GloSensor cAMP DNA construct overnight and grown in Poly-L-Lys-coated 384-well plates in DMEM + 1% FBS at a density of 15–20K cells in a volume of 40 μl per well for GloSensor cAMP assay. After 6 h or overnight recovery, cells were removed from culture medium and received 20 μl/well buffer C (20 mM HEPES, 1x HBSS, pH 7.40), 10 μl different concentrations of nalfurafine, 42B or U50,488H was added and incubated at room temperature for 15 min, then 10 μl of 4 mM luciferin supplemented with isoproternol at a final concentration of 200 nM were added and the luminescence counter was used to record chemiluminescence in relative luminescence units after 15 minutes. For Tango assay, HEK293 cells stably expressing a tTA-dependent luciferase reporter and a β-arrestin2-TEV fusion gene were transfected with the human KOR overnight80, 81 and plated in DMEM supplemented + 1% FBS in Poly-L-Lys-coated 384-well plates at a density of 15,000 cells in a volume of 40 μl per well. After 6 h or overnight recovery, each well was treated with different concentrations of nalfurafine, 42B or U50,488H dissolved in 10 μl buffer C overnight. Then, medium and drug solutions were removed and 20 μl per well of BrightGlo reagent (diluted 20-fold with buffer C, Promega, Madison, WI) was added. Cells were incubated for 20 min at room temperature in the dark before being counted on a luminescence counter. Both GloSensor cAMP and Tango assay were performed triplicate three times. EC50, Emax and RAi of each agonist were calculated from the same concentration-response curves with the method developed and refined by Ehlert and colleagues25–27. The difference between the log RAi values of an agonist for the GloSensor cAMP (log RAi−G) and Tango (log RAi−b) assays was calculated as a measure of the log agonist-bias [log (RAi–G/RAi−b)]. For each agonist, RAi−G was divided by RAi−b to estimate the bias factor to G protein.
Statistical analysis
Statistical analysis was performed using GraphPad Prism version 8.2.1 (GraphPad Software Inc., La Jolla, CA). All data are expressed as the mean ± standard error of the mean. One- or two-way analysis of variance or t-tests were used. Individual group comparisons were performed with Dunnett’s, Tukey’s or Sidak’s post-hoc test. P < 0.05 was considered to indicate a statistically significant difference.
Supplementary Material
Acknowledgment:
We thank Psychoactive Drug Screening Program of the National Institute of Mental Health, which is directed by Dr. Bryan Roth of University of North Carolina, for performing GloSensor and Tango assays. We are grateful to Dr. Xi-Ping Huang in Dr. Roth’s lab who performed the assays. Alex Willhouse’s technical help is greatly appreciated.
Funding Sources:
This work was supported by NIH grants R01 DA041359, R21 DA045274 and P30 DA013429 (L.-Y.L-C) and R01 DA024022, R01DA044855 and UG3DA050311 (YZ).
Abbreviations
- CPA
conditioned place aversion
- CPP
conditioned place preference
- DOR
δ opioid receptor
- KOR
κ opioid receptor
- MOR
μ opioid receptor
- NOR
nociceptin / orphanin FQ receptor
- norBNI
norbinaltorphimine
- RAi
intrinsic reactive activity
References
- 1.von Voigtlander PF, Lahti RA, and Ludens JH (1983) U-50,488: a selective and structurally novel non-Mu (kappa) opioid agonist, J. Pharmacol. Exp. Ther 224, 7–12. [PubMed] [Google Scholar]
- 2.Pande AC, Pyke RE, Greiner M, Wideman GL, Benjamin R, and Pierce MW (1996) Analgesic efficacy of enadoline versus placebo or morphine in postsurgical pain, Clin. Neuropharmacol 19, 451–456. [DOI] [PubMed] [Google Scholar]
- 3.Cowan A, Kehner GB, and Inan S (2015) Targeting itch with ligands selective for kappa opioid receptors, Handb. Exp. Pharmacol 226, 291–314. [DOI] [PubMed] [Google Scholar]
- 4.Pfeiffer A, Brantl V, Herz A, and Emrich HM (1986) Psychotomimesis mediated by kappa opiate receptors, Science 233, 774–776. [DOI] [PubMed] [Google Scholar]
- 5.Ranganathan M, Schnakenberg A, Skosnik PD, Cohen BM, Pittman B, Sewell RA, and D’Souza DC (2012) Dose-related behavioral, subjective, endocrine, and psychophysiological effects of the kappa opioid agonist Salvinorin A in humans, Biol. Psychiatry 72, 871–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nakao K, and Mochizuki H (2009) Nalfurafine hydrochloride: a new drug for the treatment of uremic pruritus in hemodialysis patients, Drugs Today (Barc.) 45, 323–329. [DOI] [PubMed] [Google Scholar]
- 7.Kumagai H, Ebata T, Takamori K, Miyasato K, Muramatsu T, Nakamoto H, Kurihara M, Yanagita T, and Suzuki H (2012) Efficacy and safety of a novel k-agonist for managing intractable pruritus in dialysis patients, Am. J. Nephrol 36, 175–183. [DOI] [PubMed] [Google Scholar]
- 8.Kumagai H, Ebata T, Takamori K, Muramatsu T, Nakamoto H, and Suzuki H (2010) Effect of a novel kappa-receptor agonist, nalfurafine hydrochloride, on severe itch in 337 haemodialysis patients: a Phase III, randomized, double-blind, placebo-controlled study, Nephrol. Dial. Transplant 25, 1251–1257. [DOI] [PubMed] [Google Scholar]
- 9.Kozono H, Yoshitani H, and Nakano R (2018) Post-marketing surveillance study of the safety and efficacy of nalfurafine hydrochloride (Remitch((R)) capsules 2.5 mug) in 3,762 hemodialysis patients with intractable pruritus, Int. J. Nephrol. Renovasc. Dis 11, 9–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kumada H, Miyakawa H, Muramatsu T, Ando N, Oh T, Takamori K, and Nakamoto H (2017) Efficacy of nalfurafine hydrochloride in patients with chronic liver disease with refractory pruritus: A randomized, double-blind trial, Hepatol. Res 47, 972–982. [DOI] [PubMed] [Google Scholar]
- 11.Liu JJ, Chiu YT, DiMattio KM, Chen C, Huang P, Gentile TA, Muschamp JW, Cowan A, Mann M, and Liu-Chen LY (2019) Phosphoproteomic approach for agonist-specific signaling in mouse brains: mTOR pathway is involved in kappa opioid aversion, Neuropsychopharmacology 44, 939–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kaski SW, White AN, Gross JD, Trexler KR, Wix K, Harland AA, Prisinzano TE, Aube J, Kinsey SG, Kenakin T, Siderovski DP, and Setola V (2019) Preclinical testing of nalfurafine as an opioid-sparing adjuvant that potentiates analgesia by the mu opioid receptor-targeting agonist morphine, J. Pharmacol. Exp. Ther 371, 487–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Townsend EA, Naylor JE, Negus SS, Edwards SR, Qureshi HN, McLendon HW, McCurdy CR, Kapanda CN, do Carmo JM, da Silva FS, Hall JE, Sufka KJ, and Freeman KB (2017) Effects of nalfurafine on the reinforcing, thermal antinociceptive, and respiratory-depressant effects of oxycodone: modeling an abuse-deterrent opioid analgesic in rats, Psychopharmacology (Berl) 234, 2597–2605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zamarripa CA, Naylor JE, Huskinson SL, Townsend EA, Prisinzano TE, and Freeman KB (2020) Kappa opioid agonists reduce oxycodone self-administration in male rhesus monkeys, Psychopharmacology (Berl) 237, 1471–1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhou Y, and Kreek MJ (2019) Clinically utilized kappa-opioid receptor agonist nalfurafine combined with low-dose naltrexone prevents alcohol relapse-like drinking in male and female mice, Brain Res. 1724, 146410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhou Y, and Kreek MJ (2019) Combination of Clinically Utilized Kappa-Opioid Receptor Agonist Nalfurafine With Low-Dose Naltrexone Reduces Excessive Alcohol Drinking in Male and Female Mice, Alcohol. Clin. Exp. Res 43, 1077–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang Y, Tang K, Inan S, Siebert D, Holzgrabe U, Lee DY, Huang P, Li JG, Cowan A, and Liu-Chen L-Y (2005) Comparison of pharmacological activities of three distinct k ligands (Salvinorin A, TRK-820 and 3FLB) on k opioid receptors in vitro and their antipruritic and antinociceptive activities in vivo., J. Pharmacol. Exp. Ther 312, 220–230. [DOI] [PubMed] [Google Scholar]
- 18.Endoh T, Matsuura H, Tajima A, Izumimoto N, Tajima C, Suzuki T, Saitoh A, Suzuki T, Narita M, Tseng L, and Nagase H (1999) Potent antinociceptive effects of TRK-820, a novel kappa-opioid receptor agonist, Life Sci. 65, 1685–1694. [DOI] [PubMed] [Google Scholar]
- 19.Endoh T, Tajima A, Suzuki T, Kamei J, Narita M, Tseng L, and Nagase H (2000) Characterization of the antinociceptive effects of TRK-820 in the rat, Eur. J. Pharmacol 387, 133–140. [DOI] [PubMed] [Google Scholar]
- 20.Endoh T, Tajima A, Izumimoto N, Suzuki T, Saitoh A, Suzuki T, Narita M, Kamei J, Tseng LF, Mizoguchi H, and Nagase H (2001) TRK-820, a selective kappa-opioid agonist, produces potent antinociception in cynomolgus monkeys, Jpn. J. Pharmacol 85, 282–290. [DOI] [PubMed] [Google Scholar]
- 21.Umeuchi H, Togashi Y, Honda T, Nakao K, Okano K, Tanaka T, and Nagase H (2003) Involvement of central mu-opioid system in the scratching behavior in mice, and the suppression of it by the activation of kappa-opioid system, Eur. J. Pharmacol 477, 29–35. [DOI] [PubMed] [Google Scholar]
- 22.Inan S, Dun NJ, and Cowan A (2011) Investigation of gastrin-releasing peptide as a mediator for 5’-guanidinonaltrindole-induced compulsive scratching in mice, Peptides 32, 286–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nagase H, and Fujii H (2013) Essential structure of the kappa opioid receptor agonist nalfurafine for binding to the kappa receptor, Curr. Pharm. Des 19, 7400–7414. [DOI] [PubMed] [Google Scholar]
- 24.Che T, Majumdar S, Zaidi SA, Ondachi P, McCorvy JD, Wang S, Mosier PD, Uprety R, Vardy E, Krumm BE, Han GW, Lee MY, Pardon E, Steyaert J, Huang XP, Strachan RT, Tribo AR, Pasternak GW, Carroll FI, Stevens RC, Cherezov V, Katritch V, Wacker D, and Roth BL (2018) Structure of the Nanobody-Stabilized Active State of the Kappa Opioid Receptor, Cell 172, 55–67 e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.DiMattio KM, Ehlert FJ, and Liu-Chen LY (2015) Intrinsic relative activities of kappa opioid agonists in activating Galpha proteins and internalizing receptor: Differences between human and mouse receptors, Eur. J. Pharmacol 761, 235–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Griffin MT, Figueroa KW, Liller S, and Ehlert FJ (2007) Estimation of agonist activity at G protein-coupled receptors: analysis of M2 muscarinic receptor signaling through Gi/o,Gs, and G15, J. Pharmacol. Exp. Ther 321, 1193–1207. [DOI] [PubMed] [Google Scholar]
- 27.Tran JA, Chang A, Matsui M, and Ehlert FJ (2009) Estimation of relative microscopic affinity constants of agonists for the active state of the receptor in functional studies on M2 and M3 muscarinic receptors, Mol. Pharmacol 75, 381–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Schattauer SS, Kuhar JR, Song A, and Chavkin C (2017) Nalfurafine is a G-protein biased agonist having significantly greater bias at the human than rodent form of the kappa opioid receptor, Cell. Signal 32, 59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dunn AD, Reed B, Erazo J, Ben-Ezra A, and Kreek MJ (2019) Signaling Properties of Structurally Diverse Kappa Opioid Receptor Ligands: Toward in Vitro Models of in Vivo Responses, ACS Chem. Neurosci 10, 3590–3600. [DOI] [PubMed] [Google Scholar]
- 30.Rajagopal S, Ahn S, Rominger DH, Gowen-MacDonald W, Lam CM, DeWire SM, Violin JD, and Lefkowitz RJ (2011) Quantifying ligand bias at seven-transmembrane receptors, Mol. Pharmacol 80, 367–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kenakin T (2015) Gaddum Memorial Lecture 2014: receptors as an evolving concept: from switches to biased microprocessors, Br. J. Pharmacol 172, 4238–4253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Klein Herenbrink C, Sykes DA, Donthamsetti P, Canals M, Coudrat T, Shonberg J, Scammells PJ, Capuano B, Sexton PM, Charlton SJ, Javitch JA, Christopoulos A, and Lane JR (2016) The role of kinetic context in apparent biased agonism at GPCRs, Nat. Commun 7, 10842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lane JR, May LT, Parton RG, Sexton PM, and Christopoulos A (2017) A kinetic view of GPCR allostery and biased agonism, Nat. Chem. Biol 13, 929–937. [DOI] [PubMed] [Google Scholar]
- 34.Gillis A, Gondin AB, Kliewer A, Sanchez J, Lim HD, Alamein C, Manandhar P, Santiago M, Fritzwanker S, Schmiedel F, Katte TA, Reekie T, Grimsey NL, Kassiou M, Kellam B, Krasel C, Halls ML, Connor M, Lane JR, Schulz S, Christie MJ, and Canals M (2020) Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists, Sci. Signal 13, eaaz3140. [DOI] [PubMed] [Google Scholar]
- 35.Huang W, Manglik A, Venkatakrishnan AJ, Laeremans T, Feinberg EN, Sanborn AL, Kato HE, Livingston KE, Thorsen TS, Kling RC, Granier S, Gmeiner P, Husbands SM, Traynor JR, Weis WI, Steyaert J, Dror RO, and Kobilka BK (2015) Structural insights into micro-opioid receptor activation, Nature 524, 315–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Murray CW, and Cowan A (1991) Tonic pain perception in the mouse: differential modulation by three receptor-selective opioid agonists, J. Pharmacol. Exp. Ther 257, 335–341. [PubMed] [Google Scholar]
- 37.White KL, Robinson JE, Zhu H, DiBerto JF, Polepally PR, Zjawiony JK, Nichols DE, Malanga CJ, and Roth BL (2015) The G protein-biased kappa-opioid receptor agonist RB-64 Is analgesic with a unique spectrum of activities in vivo, J. Pharmacol. Exp. Ther 352, 98–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mucha RF, and Herz A (1985) Motivational properties of kappa and mu opioid receptor agonists studied with place and taste preference conditioning, Psychopharmacology (Berl) 86, 274–280. [DOI] [PubMed] [Google Scholar]
- 39.Shippenberg TS, and Herz A (1986) Differential effects of mu and kappa opioid systems on motivational processes, NIDA Res. Monogr 75, 563–566. [PubMed] [Google Scholar]
- 40.Tsuji M, Takeda H, Matsumiya T, Nagase H, Narita M, and Suzuki T (2001) The novel kappa-opioid receptor agonist TRK-820 suppresses the rewarding and locomotor-enhancing effects of morphine in mice, Life Sci. 68, 1717–1725. [DOI] [PubMed] [Google Scholar]
- 41.Takemori AE, Ho BY, Naeseth JS, and Portoghese PS (1988) Nor-binaltorphimine, a highly selective kappa-opioid antagonist in analgesic and receptor binding assays, J. Pharmacol. Exp. Ther 246, 255–258. [PubMed] [Google Scholar]
- 42.Endoh T, Matsuura H, Tanaka C, and Nagase H (1992) Nor-binaltorphimine: a potent and selective kappa-opioid receptor antagonist with long-lasting activity in vivo, Arch. Int. Pharmacodyn. Ther 316, 30–42. [PubMed] [Google Scholar]
- 43.Broadbear JH, Negus SS, Butelman ER, De Costa BR, and Woods JH (1994) Differential effects of systemically administered nor-binaltorphimine (nor-BNI) on kappa-opioid agonists in the mouse writhing assay, Psychopharmacology (Berl) 115, 311–319. [DOI] [PubMed] [Google Scholar]
- 44.Yamamoto A, and Sugimoto Y (2010) Involvement of peripheral mu opioid receptors in scratching behavior in mice, Eur. J. Pharmacol 649, 336–341. [DOI] [PubMed] [Google Scholar]
- 45.Mague SD, Isiegas C, Huang P, Liu-Chen LY, Lerman C, and Blendy JA (2009) Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior, Proc. Natl. Acad. Sci. U. S. A 106, 10847–10852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.von Voigtlander PF, and Lewis RA (1988) Analgesic and mechanistic evaluation of spiradoline, a potent kappa opioid, J. Pharmacol. Exp. Ther 246, 259–262. [PubMed] [Google Scholar]
- 47.Kitchen I, and Rowan KM (1984) Differences in the effects of mu- and delta-opioid receptor antagonists upon plasma corticosterone levels in stressed mice, Eur. J. Pharmacol 101, 153–156. [DOI] [PubMed] [Google Scholar]
- 48.Nakamura A, Yasufuku K, Shimada S, Aritomi H, Furue Y, Chiba H, Muramoto M, Takase K, Koike K, Matsumoto T, Shimada T, Watari R, Matsuzaki T, Asaki T, Kanemasa T, and Fujita M (2020) The antagonistic activity profile of naloxone in μ-opioid receptor agonist-induced psychological dependence, Neurosci. Lett 735, 135177. [DOI] [PubMed] [Google Scholar]
- 49.Khroyan TV, Polgar WE, Orduna J, Montenegro J, Jiang F, Zaveri NT, and Toll L (2011) Differential effects of nociceptin/orphanin FQ (NOP) receptor agonists in acute versus chronic pain: studies with bifunctional NOP/μ receptor agonists in the sciatic nerve ligation chronic pain model in mice, J. Pharmacol. Exp. Ther 339, 687–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Morgenweck J, Frankowski KJ, Prisinzano TE, Aube J, and Bohn LM (2015) Investigation of the role of betaarrestin2 in kappa opioid receptor modulation in a mouse model of pruritus, Neuropharmacology 99, 600–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bruchas MR, Macey TA, Lowe JD, and Chavkin C (2006) Kappa opioid receptor activation of p38 MAPK is GRK3- and arrestin-dependent in neurons and astrocytes., J. Biol. Chem 281, 18081–18089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bruchas MR, Schindler AG, Shankar H, Messinger DI, Miyatake M, Land BB, Lemos JC, Hagan CE, Neumaier JF, Quintana A, Palmiter RD, and Chavkin C (2011) Selective p38alpha MAPK deletion in serotonergic neurons produces stress resilience in models of depression and addiction, Neuron 71, 498–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bruchas MR, Land BB, Aita M, Xu M, Barot SK, Li S, and Chavkin C (2007) Stress-induced p38 mitogen-activated protein kinase activation mediates kappa-opioid-dependent dysphoria, J. Neurosci 27, 11614–11623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ehrich JM, Messinger DI, Knakal CR, Kuhar JR, Schattauer SS, Bruchas MR, Zweifel LS, Kieffer BL, Phillips PE, and Chavkin C (2015) Kappa opioid receptor-induced aversion requires p38 MAPK Aactivation in VTA dopamine neurons, J. Neurosci 35, 12917–12931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhou L, Lovell KM, Frankowski KJ, Slauson SR, Phillips AM, Streicher JM, Stahl E, Schmid CL, Hodder P, Madoux F, Cameron MD, Prisinzano TE, Aube J, and Bohn LM (2013) Development of functionally selective, small molecule agonists at kappa opioid receptors, J. Biol. Chem 288, 36703–36716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Brust TF, Morgenweck J, Kim SA, Rose JH, Locke JL, Schmid CL, Zhou L, Stahl EL, Cameron MD, Scarry SM, Aube J, Jones SR, Martin TJ, and Bohn LM (2016) Biased agonists of the kappa opioid receptor suppress pain and itch without causing sedation or dysphoria, Sci. Signal 9, ra117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ko MC, and Husbands SM (2009) Effects of atypical kappa-opioid receptor agonists on intrathecal morphine-induced itch and analgesia in primates, J. Pharmacol. Exp. Ther 328, 193–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ando A, Sasago S, Ohzone Y, and Miyamoto Y (2016) Drug-Drug Interactions of a Novel kappa-Opioid Receptor Agonist, Nalfurafine Hydrochloride, Involving the P-Glycoprotein, Eur. J. Drug Metab. Pharmacokinet 41, 549–558. [DOI] [PubMed] [Google Scholar]
- 59.Ando A, Oshida K, Fukuyama S, Watanabe A, Hashimoto H, and Miyamoto Y (2012) Identification of human cytochrome P450 enzymes involved in the metabolism of a novel к-opioid receptor agonist, nalfurafine hydrochloride, Biopharm. Drug Dispos 33, 257–264. [DOI] [PubMed] [Google Scholar]
- 60.Chen Y, Mestek A, Liu J, Hurley JA, and Yu L (1993) Molecular cloning and functional expression of a m-opioid receptor from rat brain, Mol. Pharmacol 44, 8–12. [PubMed] [Google Scholar]
- 61.Evans CJ, Keith DE, Morrison H, Magendzo K, and Edwards RH (1992) Cloning of a delta opioid receptor by functional expression, Science 258, 1952–1955. [DOI] [PubMed] [Google Scholar]
- 62.Zhu J, Chen C, Xue JC, Kunapuli S, DeRiel JK, and Liu-Chen L-Y (1995) Cloning of a human kappa opioid receptor from the brain, Life Sci. 56, L201–L207. [DOI] [PubMed] [Google Scholar]
- 63.Adapa ID, and Toll L (1997) Relationship between binding affinity and functional activity of nociceptin/orphanin FQ, Neuropeptides 31, 403–408. [DOI] [PubMed] [Google Scholar]
- 64.Zhu J, Luo L-Y, Chen C, and Liu-Chen L-Y (1997) Activation of the cloned human k opioid receptor by agonists enhances [35S]GTPgS binding to membranes: Determination of potencies and efficacies of ligands, J. Pharmacol. Exp. Ther 282, 676–684. [PubMed] [Google Scholar]
- 65.Chen C, Xue JC, Zhu J, Chen YW, Kunapuli S, de Riel JK, Yu L, and Liu-Chen L-Y (1995) Characterization of irreversible binding of beta-funaltrexamine to the cloned rat mu opioid receptor, J. Biol. Chem 270, 17866–17870. [DOI] [PubMed] [Google Scholar]
- 66.Huang P, Kehner GB, Cowan A, and Liu-Chen L-Y (2001) Comparison of pharmacological activities of buprenorphine and norbuprenorphine: norbuprenorphine is a potent opioid agonist, J. Pharmacol. Exp. Ther 297, 688–695. [PubMed] [Google Scholar]
- 67.Stefanucci A, Dimmito MP, Macedonio G, Ciarlo L, Pieretti S, Novellino E, Lei W, Barlow D, Houseknecht KL, Streicher JM, and Mollica A (2020) Potent, Efficacious, and Stable Cyclic Opioid Peptides with Long Lasting Antinociceptive Effect after Peripheral Administration, J. Med. Chem 63, 2673–2687. [DOI] [PubMed] [Google Scholar]
- 68.Roberts BC, and Mancera RL (2008) Ligand-protein docking with water molecules, J. Chem. Inf. Model 48, 397–408. [DOI] [PubMed] [Google Scholar]
- 69.Kumar A, and Zhang KY (2013) Investigation on the effect of key water molecules on docking performance in CSARdock exercise, J. Chem. Inf. Model 53, 1880–1892. [DOI] [PubMed] [Google Scholar]
- 70.Verdonk ML, Chessari G, Cole JC, Hartshorn MJ, Murray CW, Nissink JW, Taylor RD, and Taylor R (2005) Modeling water molecules in protein-ligand docking using GOLD, J. Med. Chem 48, 6504–6515. [DOI] [PubMed] [Google Scholar]
- 71.Ballesteros JA, and Weinstein H (1995) Integrated methods for the construction of three-dimensional models and computational probing of structure-function relations in G protein-coupled receptors. In Methods in Neurosciences pp 366–428 Elsevier. [Google Scholar]
- 72.Manglik A, Kruse AC, Kobilka TS, Thian FS, Mathiesen JM, Sunahara RK, Pardo L, Weis WI, Kobilka BK, and Granier S (2012) Crystal structure of the micro-opioid receptor bound to a morphinan antagonist, Nature 485, 321–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wu H, Wacker D, Mileni M, Katritch V, Han GW, Vardy E, Liu W, Thompson AA, Huang XP, Carroll FI, Mascarella SW, Westkaemper RB, Mosier PD, Roth BL, Cherezov V, and Stevens RC (2012) Structure of the human kappa-opioid receptor in complex with JDTic, Nature 485, 327–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wang H, Zaidi SA, and Zhang Y (2017) Binding mode analyses of NAP derivatives as mu opioid receptor selective ligands through docking studies and molecular dynamics simulation, Bioorg. Med. Chem 25, 2463–2471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zheng Y, Obeng S, Wang H, Stevens DL, Komla E, Selley DE, Dewey WL, Akbarali HI, and Zhang Y (2018) Methylation Products of 6β- N-Heterocyclic Substituted Naltrexamine Derivatives as Potential Peripheral Opioid Receptor Modulators, ACS Chem. Neurosci 9, 3028–3037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zheng Y, Obeng S, Wang H, Jali AM, Peddibhotla B, Williams DA, Zou C, Stevens DL, Dewey WL, Akbarali HI, Selley DE, and Zhang Y (2019) Design, Synthesis, and Biological Evaluation of the Third Generation 17-Cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(4’-pyridyl)carboxamido]morphinan (NAP) Derivatives as μ/κ Opioid Receptor Dual Selective Ligands, J. Med. Chem 62, 561–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Obeng S, Wang H, Jali A, Stevens DL, Akbarali HI, Dewey WL, Selley DE, and Zhang Y (2019) Structure-Activity Relationship Studies of 6alpha- and 6beta-Indolylacetamidonaltrexamine Derivatives as Bitopic Mu Opioid Receptor Modulators and Elaboration of the “Message-Address Concept” To Comprehend Their Functional Conversion, ACS Chem. Neurosci 10, 1075–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Murray CW, Porreca F, and Cowan A (1988) Methodological refinements to the mouse paw formalin test. An animal model of tonic pain, J. Pharmacol. Methods 20, 175–186. [DOI] [PubMed] [Google Scholar]
- 79.Xu W, Wang Y, Ma Z, Chiu YT, Huang P, Rasakham K, Unterwald E, Lee DY, and Liu-Chen LY (2013) l-Isocorypalmine reduces behavioral sensitization and rewarding effects of cocaine in mice by acting on dopamine receptors, Drug Alcohol Depend. 133, 693–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Barnea G, Strapps W, Herrada G, Berman Y, Ong J, Kloss B, Axel R, and Lee KJ (2008) The genetic design of signaling cascades to record receptor activation, Proc. Natl. Acad. Sci. U. S. A 105, 64–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kroeze WK, Sassano MF, Huang XP, Lansu K, McCorvy JD, Giguere PM, Sciaky N, and Roth BL (2015) PRESTO-Tango as an open-source resource for interrogation of the druggable human GPCRome, Nat. Struct. Mol. Biol 22, 362–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.