Abstract
Mitragynine, an alkaloid present in the leaves of Mitragyna speciosa (kratom), has a complex pharmacology that includes low efficacy agonism at μ-opioid receptors (MORs). This study examined the activity of mitragynine at adrenergic α2 receptors (Aα2Rs) in vitro and in vivo. Mitragynine displaced a radiolabeled Aα2R antagonist ([3H] RX821002) from human Aα2ARs in vitro with lower affinity (Ki = 1260 nM) than the agonists (−)-epinephrine (Ki = 263 nM) or lofexidine (Ki = 7.42 nM). Mitragynine did not significantly stimulate [35S]GTPγS binding at Aα2ARs in vitro, but in rats trained to discriminate 32 mg/kg mitragynine from vehicle (intraperitoneally administered; i.p.), mitragynine exerted an Aα2R agonist-like effect. Both α2R antagonists (atipamezole and yohimbine) and MOR antagonists (naloxone and naltrexone) produced rightward shifts in mitragynine discrimination dose-effect function and Aα2R agonists lofexidine and clonidine produced leftward shifts. In the mitragynine trained rats, Aα2R agonists also produced leftward shifts in discrimination dose-effect functions for morphine and fentanyl. In a separate rat cohort trained to discriminate 3.2 mg/kg i.p. morphine from vehicle, naltrexone produced a rightward shift, but neither an Aα2R agonist or antagonist affected morphine discrimination. In a hypothermia assay, both lofexidine and clonidine produced marked effects antagonized by yohimbine. Mitragynine did not produce hypothermia. Together, these data demonstrate that mitragynine acts in vivo like an Aα2R agonist, although its failure to induce hypothermia or stimulate [35S]GTPγS binding in vitro, suggests that mitragynine maybe a low efficacy Aα2R agonist.
Keywords: Adrenergic α2 receptor, Kratom, Mitragynine, Opioid
1. Introduction
Opioid overdose is a leading cause of death for individuals under age 50 in the United States (US) and has decreased US life expectancy (Butelman et al., 2023; Crimmins and Zhang, 2019). Current medications approved by the US Food and Drug Administration (FDA) for opioid use disorder (OUD) fall into two pharmacological categories: μ-opioid receptor (MOR) and adrenergic-α2 receptor (Aα2R) ligands. Methadone and buprenorphine (MOR agonists) are both approved to treat opioid withdrawal symptoms and used to prevent relapse, although these MOR agonists have abuse liabilities (Hiranita et al., 2014; Timko et al., 2016). The Aα2R agonist lofexidine, is indicated for short-term use to alleviate opioid withdrawal symptoms during the initial phase of detoxification. In general, Aα2R agonists including off label use of clonidine are effective treatments of opioid withdrawal symptoms (Doughty et al., 2019; Gold et al., 1979).
Mitragyna speciosa (kratom), a plant native to Southeast Asia, is used as a short- or longer-term substitute for opioids (Garcia-Romeu et al., 2020; Singh et al., 2022; Vicknasingam et al., 2010). Mitragynine is the most abundant kratom alkaloid, i.e., ~40% of the total alkaloid content (Chear et al., 2021; Hassan et al., 2013), is a low efficacy MOR agonist (Váradi et al., 2016), and produces antinociception in mice (Macko et al., 1972) that is blocked by the MOR antagonist naloxone (Matsumoto et al., 1996b) and is absent in transgenic mice lacking MORs (Kruegel et al., 2019). In rats discriminating morphine, naltrexone blocks the capacity of mitragynine to exert morphine-like discriminative stimulus effects (Harun et al., 2015; Obeng et al., 2021) but not mitragynine-induced decreases in food-maintained responding; suggesting that some of mitragynine’s effects are not MOR mediated (Hiranita et al., 2019; Obeng et al., 2021).
In vitro, mitragynine displaces tritiated Aα2R antagonists with a 10-fold lower affinity than its affinity for MORs (Ellis et al., 2020; Obeng et al., 2020). In vivo, the antinociceptive and anti-allodynic effects of mitragynine are blocked by Aα2R antagonists (Farkas et al., 2022; Foss et al., 2020; Matsumoto et al., 1996a) suggesting that mitragynine might be an Aα2R agonist. These preclinical data raise the possibility that mitragynine exerts agonist activity not only at MORs but also at Aα2Rs, thereby representing a novel and valuable pharmacological profile for treatment of OUD. The current study determined the in vitro pharmacology of mitragynine at Aα2R with affinity and intrinsic efficacy assessments using competition binding and [35S]GTPγS binding assays. Sprague-Dawley rats were trained to discriminate mitragynine and we determined whether rats could distinguish between mitragynine and the Aα2R agonist lofexidine (alone and in combination with mitragynine) and examined the effects of the Aα2R selective antagonist atipamezole on mitragynine discrimination. For comparison we examined whether morphine or the MOR antagonist naltrexone substituted for mitragynine. A control cohort of rats were also trained to discriminate a dose of morphine; the effects of Aα2R ligands on morphine discrimination were examined. Finally, the hypothermic and antinociceptive effects of Aα2R and MOR agonism were assessed (Minor et al., 1989; Obeng et al., 2022).
2. Materials and methods
2.1. Compounds
Salt and enantiomeric forms of the following drugs used were: [3H] RX821002 (PerkinElmer, Boston, MA), [35S]GTPγS (PerkinElmer), atipamezole hydrochloride (HCl: Antisedan®, Zoetis U.S., Durham, NC), clonidine HCl (Duraclon®, Mylan N.V., Canonsburg, PA), (−)-epinephrine (Sigma-Aldrich Co., St. Louis, MO), fentanyl HCl (National Institute on Drug Abuse, Drug Supply Program, Rockville, MD), lofexidine HCl (Sigma-Aldrich Co.), (−)-mitragynine HCl [extracted as published (Maxwell et al., 2020)], (−)-morphine sulfate (National Institute on Drug Abuse), (−)-naltrexone HCl (Sigma-Aldrich Co.), (−)-naloxone HCl dihydrate (Sigma-Aldrich Co.), and yohimbine HCl (Sigma-Aldrich Co.). (−)-Mitragynine HCl was available in our alkaloid library and was isolated and structural elucidated through 1H NMR, 13C NMR, and HRMS using Bruker model AMX 500 and Avance NEO 600 NMR spectrometers operating at 500 and 600 MHz in 1H and 126 and 151 MHz in 13C, respectively. HRMS and purity (≥95%) were determined using an Agilent 1290 Infinity series ultraperformance liquid chromatography (UPLC) system equipped with photodiode array detector and quadrupole-time-of-flight (QTOF) Agilent 6540 mass spectrometer. We have previously reported all spectra data for (−)-mitragynine HCl (Obeng et al., 2020). Doses/concentrations are expressed as the weight of the salt form listed above, or as a base if no salt form is noted. For the in vitro studies, compounds were dissolved in dimethyl sulfoxide (Sigma-Aldrich Co.) to form 10 mM stock concentrations. For the in vivo studies, the vehicle consisted of sterile water containing 5% Tween 80 (polyoxyethylenesorbitanmonooleate, Sigma-Aldrich Co.) and 5% propylene glycol (Sigma-Aldrich Co.); each solution was filtered with a 0.2-μm pore size syringe filter (Millex-LG, 0.20 μm, SLLG025SS), and compound doses and vehicle were administered intraperitoneally (i.p.) in a volume of 1.0 mL/kg of body weight except mitragynine, which was prepared in volumes of 1.0–10 mL/kg due to limited solubility. Doses and pretreatment times were chosen from previous studies (Jalal et al., 2018; Obeng et al., 2020, 2021).
2.2. In vitro procedures
Aα2AR was selected for affinity and efficacy assays because it is the major Aα2R subtype in the brain (Bücheler et al., 2002).
2.2.1. In vitro receptor binding procedure
Monoclonal Chinese hamster ovary (CHO)–K1 cells expressed human Aα2AR (PerkinElmer). The Bradford protein assay was used to determine and adjust the concentration of protein required for the assay (Tal et al., 1985). The Kd (mean ± SEM) and Bmax (mean ± SEM) values for [3H] RX821002 were determined using a saturation assay as 0.433 ± 0.0140 nM and 6.75 ± 0.371 pmol/mg, respectively, which were comparable to previously reported values (O’Rourke et al., 1994). Ten μg of Aα2AR membrane protein was incubated with 1.0 nM [3H]RX821002 in the presence of different concentrations of test compounds in TME [50 mM Tris (Sigma-Aldrich Co.), 3.0 mM MgCl2 (Sigma-Aldrich Co.), and 0.2 mM ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid (EGTA, Sigma-Aldrich Co.), pH 7.7] buffer for 60 min at room temperature. The bound radioligand was separated by filtration using the Connectorate filtermat harvester for 96-well microplates (Inotech, Dietikon, Switzerland) and counted for radioactivity using a Microbeta2 microplate counter (PerkinElmer). Specific Aα2AR binding was determined as the difference in binding obtained in the absence and presence of 10 μM lofexidine. The assay was conducted in triplicate and repeated at least three times.
2.2.2. [35S]GTPγS functional assay
A [35S]GTPγS functional assay (Harrison and Traynor, 2003) was employed to compare the intrinsic activity of mitragynine to Aα2AR ligands. Twenty μg of the human Aα2A-CHO-K1 membrane protein was incubated with 10 μM guanosine diphosphate (GDP), 0.1 nM [35S] GTPγS, and varying concentrations of test compounds for 90 min at room temperature. To determine antagonism at the Aα2AR, test compounds were incubated together with 3.0 μM (−)-epinephrine. Nonspecific binding was determined using 40 μM unlabeled GTPγS. TME buffer (50 mM Tris-HCl, 9.0 mM MgCl2, 0.2 mM EGTA, pH 7.4) with 150 mM NaCl and 0.14% bovine serum albumin (BSA) was used to increase agonist-stimulated binding; the final volume in each well was 300 μL. Ten μM of (−)-epinephrine was included in the assay as the maximum effective concentration at the Aα2AR. After incubation, the bound radioligand was separated from the free radioligand by filtration through a GF/B glass fiber filter paper and rinsed three times with ice-cold wash buffer (50 mM Tris-HCl, pH 7.2) using the Unifilter-96 cell harvester (PerkinElmer). Radioactivity was measured using the Microbeta2 microplate scintillation counter. All assays were determined in triplicate and repeated at least three times.
2.3. In vivo procedures
2.3.1. Animals
Adult female and adult male Sprague Dawley rats (Taconic Biosciences, Germantown, NY, N = 8; 4 females and 4 males), weighing approximately 250 and 300 g upon arrival, respectively, were housed individually in a temperature- (mean ± SEM: 21.9 ± 1.9 °C) and humidity-controlled (mean ± SEM: 53 ± 14 %) vivarium with a 12-h light/dark cycle (lights on at Eastern Daylight Time 07:00 h). Reverse osmosis-purified water was available in the vivarium at all times. Free-feeding body weights were determined whilst each subject had continuous access to 2918 Teklad global 18% protein rodent diets (Envigo, Frenchtown, NJ). Weights were maintained at ≥85% of free-feeding weight by adjusting the amount of food (Dustless Precision Pellets Grain-Based Rodent Diet, Bio-Serv, Frenchtown, NJ) provided in the home cages 30 min following daily experimental sessions, in addition to the 45-mg sucrose pellets (maximum 50) that could be obtained during operant conditioning sessions. The Body Conditioning Score (Ullman-Culleré and Foltz, 1999) was maintained at ≥2.5 for each subject. Studies were approved by the Institutional Animal Care and Use Committee (IACUC) at the University of Florida (Protocol number: 201, 810,396) and were in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals, which was fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC).
2.3.2. Apparatus for in vivo studies
2.3.2.1. Hotplate.
A 1440 Analgesia Hot Plate with RS-232 Port and Software (Columbus Instruments, Columbus, OH) was surrounded by a clear acrylic cubicle with a lid. Temperature on the plate surface was maintained at 52 ± 0.1 °C.
2.3.2.2. Rectal thermometer.
An uninsulated microprobe (50313 Rat Rectal Probe, Stoelting, Wood Dale, IL) and a digital thermometer (50315 Body Temperature Thermometer, Stoelting) were used to measure rectal temperature. Veterinary ophthalmic ointment (Puralube®, Dechra Veterinary Products, Overland Park, KS) was applied to the tip of the microprobe prior to each use.
2.3.2.3. Drug discrimination.
Each operant-conditioning chamber (Model ENV-022MD; Med Associates Inc., Fairfax, VT) measured 45 cm × 63 cm with a height of 41 cm and was enclosed in a sound-attenuating cubicle equipped with a fan. On the front wall of each chamber were two retractable, 5-cm-long levers positioned 5 cm from the midline and 9 cm above the grid floor. One amber light-emitting diode (LED) was positioned above each lever. A response was registered by a downward displacement of the lever with 0.20 N force. A dispenser (Model ENV-203–20; Med Associates Inc.) delivered 45-mg sucrose pellets to a receptacle positioned between the levers and 2 cm above the floor. A house light was mounted on the upper, opposite wall. Each chamber was connected to a Dell desktop computer (Intel® Core™ i7-7700 3.60 GHz processor, 16.0 GB of RAM, Microsoft® Windows 10) by an interface (MED-SYST-8, Med Associates Inc.). Med-PC software version V (Med Associates Inc.) controlled experimental events and provided a record of responses.
2.3.3. In vivo experimental procedures
2.3.3.1. Hotplate and rectal temperature.
Experiments were conducted in the light cycle and body weights were measured daily prior to each experiment. Each subject was placed on the hotplate and response latency was measured manually using a stopwatch (Martin Stopwatch, Martin Sports, Carlstadt, NJ) by trained and experimentally blinded raters until the subject jumped, licked or shook a back paw or up to 60 s, whichever occurred first. Immediately thereafter, the 2.0-cm long microprobe was inserted for 10 s and rectal temperature was measured. Baseline values were measured sequentially for hotplate response latency and rectal temperature in this order prior to the drug discrimination test sessions. Following determination of each baseline, a dose or vehicle was administered i.p. Immediately following the drug discrimination session, hotplate response latency and rectal temperature were measured again. In naïve rats, drugs were administered cumulatively (every 15 min). Doses were increased by a quarter log unit per injection.
2.3.3.2. Drug discrimination procedures
2.3.3.2.1. Lever-response shaping.
Sessions started with the presentation of one of the retractable levers and the illumination of the LED above the presented lever. Initially, under a fixed-ratio (FR) 1 schedule of reinforcement, each downward deflection of the lever turned off the LEDs and activated the pellet dispenser for 0.1 s followed by a 0.1-s timeout period during which LEDs were turned off, the house light was illuminated, and responding had no scheduled consequences. The number of lever presses required for food delivery was increased systematically across daily sessions to a final value of FR10. For these sessions only the right or left lever was presented and the available lever was alternated across sessions. Sessions for drug discrimination training commenced after 50 reinforcers per session were delivered within 20 min for two consecutive sessions.
2.3.3.2.2. Drug discrimination training.
Subjects were divided equally into one of two training groups: morphine (3.2 mg/kg, i.p., administered 15 min prior to sessions, 4 females and 4 males) or mitragynine (32 mg/kg, i.p., administered 30 min prior to sessions, 4 females and 4 males). Following the pretreatment interval, subjects were placed into their assigned chamber. Initially, one lever was presented with illumination of its associated LED; completion of 10 responses turned off the LED, activated the pellet dispenser, and produced a 0.1-s timeout, during which the house light was illuminated and responding had no scheduled consequence. Training sessions ended after 20 min or delivery of 50 pellets, whichever occurred first. For each subject, one lever was assigned to the training dose and the other lever to vehicle (e. g., left lever paired with drug; right lever paired with vehicle); assignments were counterbalanced among subjects and remained the same for that subject. The order of drug and vehicle training followed a double-alternation sequence (i.e., …, right, left, left, right, …). After 12 training sessions under these parameters, both levers were presented, and 10 responses were required to complete the ratio. Test sessions for a subject commenced when the following criteria were met for at least four consecutive sessions: 1) a minimum of 80% of the total responses were correct; and 2) the total of incorrect responses made prior to delivery of the first reinforcer was less than ten. After the first test, sub-sequent test sessions were conducted after the criteria were satisfied for at least one drug and one vehicle training session.
2.3.3.2.3. Testing.
Test sessions were identical to the training sessions, except ten responses on either lever resulted in delivery of food and doses of test compounds administered. Dose-effect assessments of each training drug were obtained once at the beginning of the study and a second time after tests with all other drugs were completed. Doses of test compounds were administered from doses that produced less than group averages of 20% drug-appropriate responding up to doses that produced greater than or equal to group averages of 80% drug-appropriate responding, decreased response rate to less than 20% of the vehicle control per subject, were deemed potentially toxic, or could not be increased further due to solubility limitations. For the substitution and combination tests in both training groups, each test compound was injected 15 min prior to sessions except mitragynine and naltrexone, which were administered 30 min prior to sessions. The following ranges of drug doses (mg/kg) increased in one quarter log unit were assessed: atipamezole (3.2–17.8), clonidine (0.01–0.56), fentanyl (0.0056–0.32), lofexidine (0.0178–1.0), mitragynine (3.2–56), morphine (0.32–56), naloxone (17.8–100), naltrexone (17.8–100), and yohimbine (3.2–17.8). Naloxone (0.1 mg/kg) and naltrexone (0.032 mg/kg) were combined with morphine (3.2–56 mg/kg) in morphine-trained rats, and mitragynine (17.8–56 mg/kg) in mitragynine-trained rats. Atipamezole (3.2 mg/kg) and yohimbine (3.2 mg/kg) were combined with morphine (0.56–56 mg/kg) in morphine-trained rats, and mitragynine (17.8–100 mg/kg) in mitragynine-trained rats. Clonidine (0.032 mg/kg) and lofexidine (0.1 mg/kg) were combined with fentanyl (0.0056–0.32 mg/kg) and morphine (0.32–56 mg/kg) in morphine-trained rats and combined with fentanyl (0.001–0.32 mg/kg), morphine (0.056–56 mg/kg), and mitragynine (0.56–56 mg/kg) in mitragynine-trained rats.
2.4. Data analyses
2.4.1. In vitro data analyses
To calculate binding affinity, the IC50 values were determined using average values from at least three experiments conducted in triplicate and calculated using a nonlinear, least-squares regression analysis (Prism 9; GraphPad Software, Inc., San Diego, CA). IC50 values were converted to Ki values using the Cheng-Prusoff equation (Cheng and Prusoff, 1973). Percent (−)-epinephrine-stimulated [35S]GTPγS binding (Emax) was defined as [(net-stimulated binding by a test compound)/(net-stimulated binding by 10 μM (−)-epinephrine] × 100%. All data were normalized to obtain % stimulation values relative to 10 μM (−)-epinephrine. IC50 values for antagonists obtained from the functional assays were then converted to Kb values using a functional version of the Cheng-Prusoff equation (Leff and Dougall, 1993). EC50 and IC50 values were obtained using a nonlinear, least-squares regression analysis (GraphPad Prism version 9). Comparisons were considered significant at P < 0.05. A one-way analysis of variance (ANOVA) and post hoc Bonferroni t-test were used to analyze the effects of the compound concentration using SigmaPlot version 14.0 (Systat Software Inc., Chicago, IL).
2.4.2. In vivo data analyses
The percentage of drug-appropriate responding was calculated by dividing the total number of responses on the drug-appropriate lever by the total number of responses on the drug- and vehicle-appropriate levers. The rates of responding were normalized to baseline, defined as the mean rate of responding following an injection of vehicle during all the inter-test sessions. Percentage of drug-appropriate responding for a particular test was not plotted or analyzed when: 1) rates of responding were less than 20% of the control and 2) less than half of the sample size (e.g., five out of eight rats) did not produce more than or equal to 20% of the control rate of responding. The hotplate latencies were normalized to the percentage of the maximum possible effect (%MPE), using the following formula and each animal as its own control: %MPE = 100 × (post-injection latency - pre-injection baseline latency)/(maximum latency 60 s - pre-injection baseline latency). Changes in rectal temperature were calculated per animal as the test value subtracted from the averaged baseline value in °C. Baseline values for antinociceptive latency and rectal temperature were calculated as the average for all test sessions. A within-subjects design was used with a sample size of 8 per group (N = 4 per sex) per treatment. Data are expressed as mean values (±SEM) as a function of dose. Two- and three-way repeated-measures ANOVAs followed by post-hoc Bonferroni t-tests (P < 0.05) were conducted using GraphPad Prism version 9 for Windows and SigmaPlot version 14.0; factors included dose, sex, training drug, and multiple determinations of a dose-effect function. Standard linear regression (Snedecor and Cochran, 1967) determined using GraphPad Prism version 9 was used to calculate ED50 and ED−2°C values and corresponding 95% confidence intervals (95%CI) from dose-effect functions when the mean percentage of drug-appropriate responding or maximum possible effect was increased to greater than 50%, or when response rates were decreased to less than 50%. The linear portion of the dose-effect functions were used for the analysis. All data for response rates, maximum possible effects, and changes in rectal temperature were plotted and analyzed. Potency ratios, slope ratios, and corresponding 95% CIs were calculated as the ratio of ED50 or slope values calculated from the dose-effect function (Tallarida, 2002). If the 95% CIs of the ED50 and slope values did not overlap or if the 95% CIs of the potency ratio did not include 1, the potencies were considered to be significantly different. The dose-effect functions for discriminative-stimulus and rate-decreasing effects for both training drugs (mitragynine and morphine) determined at the beginning and end of the study were compared by evaluating the potency ratios; because the ED50 values were not significantly different from each other (Supplemental Figs. S1 and S2), data from the first dose-effect assessment were used for further analyses.
3. Results
3.1. Aα2R binding affinity and intrinsic activity
The binding affinity of mitragynine at the Aα2AR was compared to those of reference Aα2R ligands (Fig. 1, leftmost panel; Table 1). The reference Aα2R ligands atipamezole, clonidine, (−)-epinephrine, lofexidine, and yohimbine concentration-dependently displaced [3H] RX821002 bound at the Aα2AR. The rank order of binding affinities at the Aα2AR was yohimbine > lofexidine ≈ atipamezole > clonidine > (−)-epinephrine > mitragynine. The intrinsic activity (Emax) and potency (EC50 or Kb) of mitragynine and reference Aα2R ligands were compared to that of (−)-epinephrine (Fig. 1, middle panel; Table 1). (−)-Epinephrine was a full agonist (Emax ± SEM = 102 ± 1.95 %) at the Aα2AR whereas clonidine was a partial agonist (Emax ± SEM = 33.2 ± 4.03 %). Lofexidine had a lower intrinsic activity than previously reported (Raffa et al., 2019), but the 10.8 ± 0.88 % maximum stimulation was significantly different from vehicle (F10,52 = 3.67; P < 0.001). Mitragynine up to 100 μM and the reference Aα2AR antagonist, atipamezole and yohimbine up to 10 μM did not significantly stimulate [35S] GTPγS binding at the Aα2AR. (Fig. 1, middle panel; p < 0.05). Yohimbine, atipamezole, lofexidine, and mitragynine all substantially decreased (−)-epinephrine-stimulated [35S]GTPγS binding (Fig. 1, rightmost panel; Table 1), although mitragynine at 100 μM (highest concentration tested) did not decrease the efficacy of (−)-epinephrine by more than 50%.
Fig. 1.

Displacement of radioligand (leftmost panel), [35S]GTPγS stimulation (middle panel), and antagonism of (−)-epinephrine stimulation (rightmost panel) at Aα2AR. Abscissae: concentrations of compounds in nM (log scale). Ordinates: leftmost panel, percentage of [3H]RX821002 bound to membrane preparations; middle and rightmost panels, percentage of maximum stimulation of [35S]GTPγS binding normalized to maximum stimulation by (−)-epinephrine as 100%. In rightmost panel, varying concentrations of test compounds were co-incubated with an EC90 concentration (3.0 μM) of (−)-epinephrine. Each data point represents the mean results of three repeated experiments; vertical bars represent SEM (N = 3) from at least three independent triplicate replications per sample. Details for statistical analyses are shown in Table 1.
Table 1.
Binding affinity (Ki), potency (EC50 and Kb), and intrinsic activity (Emax) at Aα2ARs. The data are plotted in Fig. 1.
| Compound | Ki ± SEM (nM) | or Kb* ± SEM (nM) | Emax ± SEM [% of (−)-epinephrine stimulation] |
|---|---|---|---|
| Atipamezole | 7.85 ± 0.384 | 6.82 ± 1.21* | 0.623 ± 3.04 |
| Clonidine | 45.7 ± 4.16 | 37.9 ± 4.68# | 33.2 ± 4.03 |
| (−)-Epinephrine | 263 ± 27.5 | 443 ± 104# | 102 ± 1.95 |
| Lofexidine | 7.42 ± 0.512 | 37.5 ± 0.30* | 10.8 ± 0.88 |
| Mitragynine | 1260 ± 90.2 | Not Applicable | −4.88 ± 1.29 |
| Yohimbine | 1.13 ± 0.089 | 4.16 ± 0.314* | 1.85 ± 2.20 |
Not Applicable: The EC50 value of mitragynine could not be calculated because of its low intrinsic activity, and the Kb value for mitragynine could not be calculated because it produced less than 50% reduction in the intrinsic activity of 3.0 μM (−)-epinephrine. #EC50 values and *Kb values.
3.2. Effects of training drugs mitragynine and morphine alone
The baseline hotplate response latencies (seconds), baseline rectal temperatures (°C), and mean operant response rates (responses per second) after vehicle administration in mitragynine and morphine-trained rats were not significantly different. The baseline hotplate response latencies [mean (SEM) in seconds] for morphine- and mitragynine-trained rats were 6.82 (0.095) and 8.46 (0.663), respectively. The baseline rectal temperatures [mean (SEM) in °C] for morphine- and mitragynine-trained rats were 37.4 (0.06) and 37.3 (0.14), respectively. The mean operant response rates [mean (SEM) in responses per second] after vehicle administration for morphine- and mitragynine-trained rats were 1.01 (0.064) and 0.99 (0.094), respectively. There was no sex difference in each parameter (data not shown).
The ED50 and corresponding 95% CI values for discriminative stimulus, rate-decreasing, and antinociceptive effects, as well as the ED−2°C and corresponding 95% CI values for hypothermic effects of all compounds are summarized in Tables 2 and 3 and Supplemental Tables S1–S3. In morphine-trained rats, morphine dose-dependently increased morphine-lever responding and hotplate latency (antinociception), and decreased response rates (Figs. 2 and 3). Morphine did not significantly alter rectal temperature (Fig. 3; Table 2). The potency of morphine [ED50 (95% CIs) value] to increase morphine-lever responding [1.70 (0.793, 2.75) mg/kg] was 21-fold higher than its potency to produce antinociception [35.3 (26.9, 47.3) mg/kg], consistent with a higher degree of MOR-mediated intrinsic activity being required for antinociception as compared with discriminative stimulus effects (Table 2). The potency of morphine [ED50 (95% CIs) value] to decrease rates of operant responding was 13.4 (7.03, 21.2) mg/kg.
Table 2.
Potency comparisons in mg/kg of the discriminative-stimulus, rate-decreasing, antinociceptive [ED50 (95% CIs) values], and hypothermic [ED−2C° (95% CIs) values] effects of compounds in rats trained to discriminate 3.2 mg/kg morphine. The data are plotted in Figs. 2–5.
| Test drug | ED50 | ED−2°C | Potency Ratio | ||||
|---|---|---|---|---|---|---|---|
| Discrimination | Response Rate | Antinociception | Hypothermia | Rate-Decreasing/Discrimination | Antinociception/Discrimination | Hypothermia/Discrimination | |
| Atipamezole | Inactive | 12.2 (11.0, 13.7) | 14.3 (10.1, 31.9) | Not Determined | Not Applicable | Not Applicable | Not Applicable |
| Lofexidine | Inactive | 0.382 (0.246, 0.560) | Not Determined | 0.373 (0.292, 0.466) | Not Applicable | Not Applicable | Not Applicable |
| Mitragynine | 20.9 (9.17, 118) | 35.7 (32.9, 38.8) | Inactive | Not determined | 1.71 (0.280, 4.23) | Not Applicable | Not Applicable |
| Morphine | 1.70 (0.793, 2.75) Females: 1.66 (1.11, 2.40) Males: 1.77 (0.962, 2.79) |
13.4 (7.03, 21.2) Females: 11.9 (1.50, 22.7) Males: 14.8 (9.47, 21.9) |
35.3 (26.9, 47.3) Females: 36.5 (30.7, 44.1) Males: 34.1 (22.6, 52.0) |
Inactive |
7.85 (2.56, 26.7)
Females: 7.00 (2.54, 28.7) Males: 8.65 (3.45, 27.6) |
20.7 (9.80, 59.7)
Females: 21.4 (11.2, 55.6) Males: 20.0 (8.24, 65.6) |
Not Applicable |
| Morphine + 3.2 mg/kg Atipamezole | 1.96 (1.76, 2.17) | 18.7 (1.70, 36.7) | 34.5 (27.3, 45.4) | Inactive | 9.53 (0.781, 20.8) | 17.6 (12.6, 25.7) | Not Applicable |
| Morphine + 0.1 mg/kg Lofexidine | 1.61 (1.35, 4.33) | 7.17 (1.36, 14.0) | 35.2 (29.6, 42.9) | Inactive | 4.46 (0.314, 10.4) | 21.9 (6.84, 31.7) | Not Applicable |
| Morphine + 0.032 mg/kg Naltrexone | 19.9 (15.2, 37.4) | 34.1 (27.0, 45.5) Females: 52.1 (48.3, 57.1) Males: 22.5 (9.93, 37.0) |
Not determined | Inactive | 1.71 (0.721, 2.99) Females versus Males: 2.32 (1.31, 5.74) |
Not Applicable | Not Applicable |
| Naltrexone | Inactive | 62.3 (53.9, 72.4) | Inactive | Inactive | Not Applicable | Not Applicable | Not Applicable |
Each value is a combination of females and males unless otherwise noted. Potency ratios (95% CIs) are calculated by dividing the ED50 values for the rate-decreasing or antinociceptive effects or the ED−2C° values for the hypothermic effects by the ED50 values for the discriminative stimulus effects. Significant differences are in bold.
Table 3.
Potency comparisons in mg/kg of the discriminative-stimulus, rate-decreasing, antinociceptive [ED50 (95% CIs) values], and hypothermic [ED−2C° (95% CIs) values]
| Test drug | ED50 | ED−2°C | Potency Ratio | ||||
|---|---|---|---|---|---|---|---|
| Discrimination | Response Rate | Antinociception | Hypothermia | Rate-Decreasing/Discrimination | Antinociception/Discrimination | Hypothermia/Discrimination | |
| Atipamezole | Inactive | 12.8 (10.9, 15.6) | 15.1 (11.4, 24.1) | Not Determined | Not Applicable | Not Applicable | Not Applicable |
| Lofexidine | 0.182 (0.140, 0.242) | 0.223 (0.145, 0.320) | 0.573 (0.415, 0.813) | 0.305 (0.272, 0.345) | 1.22 (0.599, 2.28) | 3.13 (1.72, 5.79) | 1.67 (1.12, 2.46) |
| Mitragynine | 14.3 (8.20, 20.9) | 50.9 (44.8, 59.4) | Inactive | Inactive | 3.55 (2.15, 7.24) | Not Applicable | Not Applicable |
| Mitragynine + 3.2 mg/kg Atipamezole | Not Determined | 60.7 (47.8, 77.5) | Inactive | Inactive | Not Applicable | Not Applicable | Not Applicable |
| Mitragynine + 0.1 mg/kg Lofexidine | 2.45 (1.26, 3.87) | 16.2 (10.8, 22.1) | Not determined | 45.8 (32.4, 94.0) | 6.63 (2.78, 17.5) | Not Applicable | 18.7 (8.38, 74.6) |
| Mitragynine + 0.032 mg/kg Naltrexone | Not determined | Not determined | Inactive | Inactive | Not Applicable | Not Applicable | Not Applicable |
| Morphine | 12.7 (10.5, 15.3) | 21.3 (3.25, 40.6) | 32.9 (28.4, 38.6) | Inactive | 1.68 (0.213, 3.85) | 2.60 (1.86, 3.67) | Not Applicable |
| Morphine + 0.1 mg/kg Lofexidine | 0.546 (0.194, 0.992) | 16.0 (4.67, 30.6) | 27.8 (25.1, 30.9) | Inactive | 29.2 (7.16, 158) | 50.9 (38.5, 159) | Not Applicable |
| Naltrexone | Inactive | 97.5 (81.3, 126) | Not Determined | Inactive | Not Applicable | Not Applicable | Not Applicable |
Each value is a combination of females and males unless otherwise noted. Potency ratios (95% CIs) are calculated by dividing the ED50 values for the rate-decreasing or antinociceptive effects or the ED−2° C values for the hypothermic effects by the ED50 values for the discriminative-stimulus effects. Significant differences are in bold.
Fig. 2.

Substitution of Aα2R and MOR ligands for 3.2 mg/kg morphine (left panels) or 32 mg/kg mitragynine (right panels). Abscissae: Vehicle and compound dose in mg/kg (i.p., log scale). Ordinates: Upper panels, percentage of responses on the training drug-appropriate lever; lower panels, mean rates of responding expressed as a percentage of vehicle control. Atipamezole (squares with crosshatch), lofexidine (diamonds), and morphine (circles) were administered 15 min before sessions while mitragynine (squares) and naltrexone (circles with crosshatch) were administered 30 min before sessions. Each gray symbol indicates a significant difference from vehicle in the corresponding training group. Each point represents the mean ± SEM (N = 4 rats/sex/dose). Details for statistical analyses are shown in Tables 2 and 3.
Fig. 3.

Antinociceptive and hypothermic effects of Aα2R and MOR ligands. Abscissae: Vehicle and compound dose in mg/kg (i.p., log scale). Ordinates: Upper panels, percent maximum possible effects; lower panels, changes in rectal temperature from baseline. The left panels are from morphine-trained rats while the right panels are from mitragynine-trained rats. Atipamezole (squares with crosshatch), lofexidine (diamonds), and morphine (circles) were administered 15 min before sessions while mitragynine (squares) and naltrexone (circles with crosshatch) were administered 30 min before sessions. Gray symbols indicate significant differences from vehicle in the corresponding training group. Each point represents the mean ± SEM (N = 4 rats/sex/dose). Details for statistical analyses are shown in Tables 2 and 3.
In morphine-trained rats, mitragynine did not produce significant antinociception up to the largest dose that could be safely studied (56 mg/kg) (F1.51,9.06 = 3.76; P = 0.0731, Fig. 3, upper left panel). Mitragynine produced a maximum of 73% drug lever responses in the morphine-trained rats while morphine produced a maximum of 70% drug lever responses in the mitragynine-trained rats. The potency of mitragynine [ED50 (95% CIs) value] to increase the percentage of mitragynine-lever responding was 14.3 (8.20, 20.9) mg/kg (Fig. 2, upper right panel) and its potency to decrease rates of operant responding was 50.9 (44.8, 59.4) mg/kg (Table 3).
At the end of the study, the dose-effect functions for each training compound were re-assessed (Supplemental Figs. S1 and S2). In both morphine- (F1.81,10.9 = 1.16; P = 0.345, F2.49,14.9 = 0.996; P = 0.409, F2.27,13.6 = 0.746; P = 0.509, and F2.60,15.6 = 0.904; P = 0.449, respectively) and mitragynine-trained (F1.49,8.96 = 0.309; P = 0.680, F3.60,21.6 = 1.41; P = 0.266, F3.15,18.9 = 1.12; P = 0.369, and F2.99,17.9 = 2.77; P = 0.0717, respectively) groups, there were no significant differences between the first and second determination for the discriminative stimulus, rate-decreasing, antinociception, or hypothermia dose-effect functions. Moreover, the antinociceptive and hypothermic effects of mitragynine and morphine in drug-naïve rats were not significantly different from those in the morphine- and mitragynine-trained rats (Supplemental Fig. S3 and Supplemental Table S1). Thus, morphine produced similar antinociceptive and hypothermic effects in drug naïve rats and in trained rats that received repeated administrations of morphine. Mitragynine produced similar antinociceptive and hypothermic effects in drug naïve rats and mitragynine-trained rats, indicating that the lack of antinociceptive effects of mitragynine in the mitragynine-trained rats is not due to tolerance.
3.3. Effects of Aα2R and MOR ligands alone
Neither naltrexone (up to 100 mg/kg) nor naloxone (up to 100 mg/kg) significantly increased morphine- or mitragynine-lever responding (Fig. 2 and Supplemental Fig. S4). Both naltrexone and naloxone reduced response rates (Fig. 2 and Supplemental Fig. S4) but neither naltrexone nor naloxone significantly altered hotplate response latency or rectal temperature (Fig. 3 and Supplemental Fig. S5). In mitragynine-trained rats, the MOR agonist fentanyl produced greater than 80% mitragynine-paired lever responses, greater than 80% antinociception, and decreased response rates (rightmost panels in Supplemental Figs. S6 and S7).
In morphine-trained rats, lofexidine did not significantly increase drug-lever responding or produce antinociception up to a dose (1.0 mg/kg) that significantly decreased operant response rates (Figs. 2 and 3, left panels). Lofexidine produced marked hypothermia, with a maximum decrease of 5.0 °C (±0.42 °C SEM) (Fig. 3, lower left panel). In mitragynine-trained rats, lofexidine increased drug-lever responding up to a maximum of 77% (±16% SEM), decreased response rates, and induced hypothermia (Figs. 2 and 3, right panels). In mitragynine-trained rats, lofexidine produced significant antinociception with a maximum possible effect of 62% (±5.1% SEM) (Fig. 3, upper right panel, F2.25,13.5 = 97.0; P < 0.001). There were no significant differences in the antinociceptive effects of lofexidine in mitragynine-trained and drug naïve rats (Fig. S3 and Table S1). Clonidine increased drug-lever responding to a maximum of 9.7% in morphine-trained rats and 55% in mitragynine-trained rats; in both groups, clonidine significantly decreased operant response rates and produced significant antinociceptive and hypothermic effects (Supplemental Figs. S4 and S5, Supplemental Tables S2 and S3). Atipamezole (up to 17.8 mg/kg) and yohimbine (up to 17.8 mg/kg) did not significantly increase drug-lever responding in either morphine- or mitragynine-trained rats (Fig. 2 and Supplemental Fig. S4; Tables 2 and 3 and Supplemental Tables S2 and S3). Atipamezole and yohimbine produced significant antinociception in both groups of rats (Fig. 3 and Supplemental Fig. S5; Tables 2 and 3 and Supplemental Tables S2 and S3). Moreover, atipamezole and yohimbine produced significant hypothermia in the mitragynine-trained rats but not in morphine-trained rats, although the decreases in rectal temperature produced by atipamezole (−1.4 °C) and yohimbine (−2.6 °C) were lower than that produced by clonidine (−4.5 °C) or lofexidine (−3.8 °C). The ED50 and ED−2°C values of atipamezole and lofexidine are shown in Tables 2 and 3, while those for clonidine and yohimbine are shown in Supplemental Tables S2 and S3.
3.4. Effects of MOR or Aα2R antagonists in combination with morphine and mitragynine
Naltrexone (0.032 mg/kg) antagonized the discriminative stimulus effects of morphine, as evidenced by a significant rightward shift in the morphine dose-effect function (Fig. 4, upper leftmost panel; Table 2). Naltrexone also significantly antagonized the discriminative stimulus effects of mitragynine, as evidenced by a rightward and downward shift of the mitragynine dose-effect function (Fig. 4, upper middle panel; Table 3). Naltrexone did not significantly modify the dose-effect function of mitragynine on rates of responding but produced a 2.5-fold rightward shift in the dose-effect function of morphine on rates of responding (Fig. 4, lower middle and leftmost panels, respectively; Tables 2 and 3) and antagonized the antinociceptive effects of morphine (56 mg/kg) in morphine-trained rats (Fig. 5, upper leftmost panel; Table 2). Naloxone (0.1 mg/kg) also antagonized the discriminative stimulus effects of morphine and mitragynine (Supplemental Fig. S6; Supplemental Tables 2 and 3), and the antinociceptive effects of morphine (Supplemental Fig. S7; Supplemental Tables 2 and 3).
Fig. 4.

Effects of atipamezole, lofexidine, and naltrexone to modify the discriminative-stimulus and rate-decreasing effects of morphine and mitragynine. Abscissae: Vehicle and compound dose in mg/kg (i.p., log scale). Ordinates: Upper panels, percentage of responses on the training drug-appropriate lever; lower panels, mean rates of responding expressed as a percentage of vehicle control. The leftmost panels show data obtained in morphine-trained rats while the middle and rightmost panels show data obtained in mitragynine-trained rats. Open circles and open squares represent data for morphine and mitragynine administered alone, respectively. Squares with crosshatch, open diamonds, and circles with crosshatch represent data for morphine or mitragynine in the presence of 3.2 mg/kg atipamezole, 0.1 mg/kg lofexidine, and 0.032 mg/kg naltrexone, respectively. All compounds were administered 15 min before sessions except mitragynine and naltrexone (30 min before sessions). Gray symbols indicate significant differences from vehicle in the corresponding training compound. Each point represents the mean ± SEM (N = 4 rats/sex/dose). Details for statistical analyses are shown in Tables 2 and 3.
Fig. 5.

Effects of atipamezole, lofexidine, and naltrexone in combination with morphine or mitragynine in hotplate antinociception and rectal temperature. Abscissae: Vehicle and compound dose in mg/kg (i.p., log scale). Ordinates: Upper panels, percent antinociceptive maximum possible effects; lower panels, changes in rectal temperature from baseline. All compounds were administered 15 min before sessions except mitragynine and naltrexone (30 min before sessions). The leftmost panels show data obtained in morphine-trained rats while the middle and rightmost panels show data obtained in mitragynine-trained rats. Open circles and open squares represent data for morphine and mitragynine, respectively, administered alone. Squares with crosshatch, open diamonds, and circles with crosshatch represent data of morphine or mitragynine in the presence of 3.2 mg/kg atipamezole, 0.1 mg/kg lofexidine, and 0.032 mg/kg naltrexone. The antinociceptive and thermoregulatory effects of mitragynine in combination with the Aα2AR antagonist atipamezole (3.2 mg/kg) and MOR antagonist naltrexone (0.032 mg/kg) are not shown because mitragynine alone did not significantly alter percent antinociceptive maximum possible effects or rectal temperature. Gray symbols indicate a significant difference from vehicle in the corresponding training compound. Each point represents the mean ± SEM (N = 4 rats/sex/dose). Details for statistical analyses are shown in Tables 2 and 3.
In morphine-trained rats, neither atipamezole (3.2 mg/kg) nor yohimbine (3.2 mg/kg) significantly altered the discriminative stimulus or rate-decreasing effects of morphine (Fig. 4 and Supplemental Fig. S6, upper leftmost panels; Table 2 and Supplemental Table S2). However, both antagonists produced rightward and downward shifts in the dose-effect functions of mitragynine to produce discriminative stimulus effects (Fig. 4, upper middle panel; Supplemental Fig. S6, upper second panel from the left; Table 3 and Supplemental Table S3). However, neither atipamezole (3.2 mg/kg) nor yohimbine (3.2 mg/kg) modified the capacity of mitragynine to decrease operant response rates (Fig. 4, lower middle panel; Supplemental Fig. S6, lower second panel from the left; Table 3 and Supplemental Table S3).
3.5. Effects of Aα2R agonists on discriminative stimulus, rate-decreasing, and antinociceptive effects of morphine
In morphine-trained rats, neither lofexidine (0.1 mg/kg) nor clonidine (0.032 mg/kg) significantly altered the dose-effect function of morphine to produce discriminative stimulus effects (Fig. 4 and Supplemental Fig. S6, upper leftmost panels; Table 2 and Supplemental Table S2). However, both lofexidine and clonidine shifted leftward the dose-effect function of morphine to decrease operant response rates (Fig. 4 and Supplemental Fig. S6, lower leftmost panels; Table 2 and Supplemental Table S2). Neither lofexidine nor clonidine significantly altered morphine antinociception (Fig. 5 and Supplemental Fig. S7, upper leftmost panels; Table 2 and Supplemental Table S2).
3.6. Effects of Aα2AR agonists on discriminative stimulus and rate-decreasing effects of mitragynine
In mitragynine-trained rats, lofexidine (0.1 mg/kg) produced 5.8- and 3.1-fold leftward shifts in the dose-effect functions of mitragynine to produce discriminative stimulus and rate-decreasing effects, respectively (Fig. 4, middle panels; Table 3), while clonidine (0.032 mg/kg) produced 1.5- and 5.0-fold leftward shifts in the dose-effect functions of mitragynine on the discriminative stimulus and rate-decreasing effects, respectively (Supplemental Fig. S6, second panels from the left; Supplemental Table S3).
In mitragynine-trained rats, lofexidine (0.1 mg/kg; Fig. 4, upper rightmost panel; Table 3) and clonidine (0.032 mg/kg; Supplemental Fig. S6, upper third panel from the left; Supplemental S3) produced 23- and 15-fold leftward shifts, respectively, in the dose-effect function of morphine to increase mitragynine-lever responding. In addition, lofexidine (Fig. 4, lower rightmost panel; Table 3) and clonidine (Supplemental Fig. S6, lower third panel from the left; Supplemental S3) significantly shifted the morphine dose-effect functions for the rate-decreasing effects to the left. However, neither lofexidine (Fig. 5, upper rightmost panel; Table 3) nor clonidine (Supplemental Fig. S7, upper third panel from the left; Supplemental S3) significantly changed the morphine dose-effect functions for the antinociceptive effects. To examine the generality of the findings with Aα2R agonists in combination with morphine in mitragynine-trained rats and not in morphine-trained rats, fentanyl was tested in combination with Aα2R agonists in both groups. In mitragynine-trained rats (Supplemental Fig. S6, rightmost panels; Supplemental S3), co-administration of fentanyl with lofexidine (0.1 mg/kg) or clonidine (0.032 mg/kg) produced 7.1- and 7.7- fold leftward shifts, respectively, in the dose-effect functions of fentanyl on the discriminative stimulus effects, and 5.0- and 3.6- fold leftward shifts, respectively, in the rate-decreasing dose-effect function of fentanyl. On the contrary, neither lofexidine (0.1 mg/kg) nor clonidine (0.032 mg/kg) produced leftward shifts in the dose-effect functions of fentanyl to produce discriminative stimulus and rate-decreasing effects in morphine-trained rats (data not shown).
3.7. Comparison of effects in females and males
There were no sex differences in any effects tested. As a result, all data points plotted are a combination of females and males unless otherwise noted.
4. Discussion
In the current study, mitragynine at the human Aα2ARs exhibited low micromolar binding affinity (see also Obeng et al., 2020), and did not stimulate [35S]GTPγS binding. The drug discrimination results reported here are consistent with mitragynine exhibiting low efficacy agonism at Aα2ARs and MORs. This was evidenced not only by antagonism of mitragynine by Aα2ARs and MOR antagonists, but also by enhancement of the effects of mitragynine, morphine, and fentanyl by Aα2AR agonists in rats discriminating mitragynine. The apparent low efficacy agonism exhibited by mitragynine at both MORs and Aα2Rs may provide advantages over currently approved FDA medications for OUD inasmuch as multiple medications (e.g., buprenorphine and lofexidine) are currently required to achieve this polypharmacology.
The affinity and intrinsic activity of the reference Aα2R agonists and antagonists obtained in this study were compared to previously reported results. Aα2AR agonists clonidine and lofexidine inhibited isoproterenol-stimulated cyclic adenosine monophosphate (cAMP) production in bovine ciliary process membranes (Jin et al., 1989). The intrinsic activity of lofexidine and clonidine at human Aα2ARs were reported to have a higher maximum intrinsic activity [70 and 90%, respectively (Raffa et al., 2019)] than observed in the current study (11 and 33%). The low in vitro intrinsic activity of lofexidine observed here could be due to a lower level of Aα2AR expression/reserve in our heterologous cell model [Bmax (mean ± SEM) = 6.75 ± 0.371 pmol/mg] compared to human Aα2ARs employed by Raffa et al. (2019) and unfortunately there was no information on Bmax values in the study by Raffa et al. (2019). When a low efficacy agonist binds, the probability of resultant receptor activation is lower than for a full agonist. However, when receptor expression is high relative to the effector population, i.e. “high reserve”, such as can be found in some heterologous models, even low efficacy agonists can activate a high enough number of receptors to saturate the downstream effector. Such assay conditions obscure the differences between high and low efficacy agonists (Kelly, 2013; Niedernberg et al., 2003). Moreover, a [35S]GTPγS assay was used in the current study versus the adenylate cyclase assay previously used and adenylate cyclase has a faster turnover rate than G-proteins resulting in signal amplification (Costa et al., 1988; Newton et al., 2016). In our laboratory, mitragynine did not stimulate Aα2AR-mediated [35S]GTPγS binding, although we previously observed insignificant human MOR-mediated [35S]GTPγS binding (Obeng et al. (2021), while mitragynine’s pharmacological profile in rats discriminating morphine was consistent with low efficacy MOR agonism. In addition, we did observe mitragynine antagonism of (the full agonist) epinephrine-induced [35S]GTPγS binding in the current study consistent with mitragynine acting as a low efficacy agonist (or antagonist) in vitro.
The current results extend previous findings demonstrating that at least some of the behavioral effects of mitragynine are mediated by MORs. As reported previously in separate groups of rats trained to discriminate either morphine or mitragynine (Harun et al., 2015; Obeng et al., 2021), tests of cross-substitution demonstrated that discriminative stimulus effects partially overlapped. That is, when tested in rats discriminating mitragynine, morphine produced a maximum of 70% drug-appropriate responding. Conversely, mitragynine produced a maximum of 73% drug-appropriate responding in rats discriminating morphine. The MOR agonist fentanyl exhibited a similar profile, producing a maximum of 84% drug-appropriate responding in the mitragynine discrimination assay. These results, coupled with antagonism of the discriminative stimulus effects of mitragynine by naltrexone (Obeng et al., 2021) in the previous and present studies, support the hypothesis that mitragynine is a MOR agonist in vivo.
Lofexidine and clonidine produced significantly higher levels of drug-appropriate responding in the mitragynine discrimination assay as compared to the morphine discrimination assay. While these results may suggest greater involvement of Aα2AR in the effects of mitragynine as compared with morphine, more parsimonious explanations must be considered and ruled out. For example, increases in drug-lever responding can sometimes indicate that a discrimination assay lacks pharmacological selectivity, i.e., there is an effect of the test compound to produce responding on both drug- and vehicle-appropriate levers that is not due to shared pharmacological mechanisms between the training and test compounds. However, this explanation might be ruled out by the results obtained with other test compounds, e.g., the Aα2AR antagonists atipamezole and yohimbine produced negligible mitragynine-appropriate responding up to doses that markedly decreased rates of responding. Partial substitution of clonidine and lofexidine was accompanied by leftward shifts in the dose-response function for not only mitragynine as a training drug, but also for morphine and fentanyl in their capacity to substitute for the mitragynine discriminative stimulus. However, this enhancement of the mitragynine-like discriminative stimulus of morphine and fentanyl occurred only in the mitragynine discrimination assay, i.e., clonidine and lofexidine did not enhance the discriminative stimulus effects of morphine in the morphine discrimination assay. Selective enhancement of the mitragynine-like discriminative stimulus of morphine by clonidine and lofexidine appears to rule out pharmacokinetic interactions as the underlying cause insofar as the same enhancement would have been expected in both the morphine and mitragynine discrimination assays. Collectively, these results implicate a pharmacodynamic interpretation whereby the discriminative stimulus effects of mitragynine are mediated at least in part by Aα2AR agonism.
Pharmacological mechanisms of mitragynine at Aα2ARs were further assessed with the antagonists atipamezole and yohimbine. Both antagonists attenuated the discriminative stimulus effects of mitragynine, but not morphine. The specific antagonism by atipamezole and yohimbine occurred at doses that appear to be selective for Aα2Rs based on previous results (Obeng et al., 2020, 2022). In addition, the opioid antagonists naltrexone and naloxone attenuated the discriminative stimulus effects of mitragynine and morphine. These findings suggest that the discriminative stimulus effects of mitragynine are mediated by both Aα2Rs and MOR, whereas there is little or no evidence for the involvement of Aα2Rs in the discriminative stimulus effects of morphine. Previous studies spanning decades have consistently demonstrated that the discriminative stimulus effects of morphine are selectively mediated by MORs (Colpaert, 1978; Obeng et al., 2021; Suzuki et al., 1995; Young et al., 1992). Thus, the discriminative stimulus effects of mitragynine are due to the dual activation of the Aα2Rs and MORs which explains why both Aα2R (mitragynine-lever responding for clonidine and lofexidine were 55% and 77%, respectively) and MORs (mitragynine-lever responding for morphine and fentanyl were 70% and 84%, respectively) agonists substituted or partially substituted for mitragynine and also why the Aα2R and MOR antagonists decreased mitragynine-appropriate lever responding. There is a reasonable scientific premise that shows that MOR and α2AR agonists interact, including both anatomical and physiological relationships between the neurotransmitters associated with these receptor types (Dang et al., 2012; Jordan et al., 2003; Tan et al., 2009; Vilardaga et al., 2008). MOR and α2AR are co-localized in brain and spinal pathways that control pain processing, and under some conditions α2AR and MOR agonists act synergistically to increase the latency of reflexive behavioral responses induced by application of a noxious heat stimulus (Drasner and Fields, 1988; Ossipov et al., 1990; Stone et al., 2014). The pharmacodynamic interactions between the Aα2R and MOR could explain why lofexidine and clonidine produced leftward shifts in the dose-effect function of fentanyl and morphine to produce mitragynine-like discriminative stimulus effects but not morphine-like discriminative stimulus effects.
We have previously shown that both lofexidine and clonidine decreased rectal temperature, whereas morphine did not significantly alter rectal temperature (Obeng et al., 2022). The hypothermic effects of both lofexidine and clonidine were enhanced by 17.8 mg/kg mitragynine but antagonized by 1.0 mg/kg yohimbine (Supplemental Fig. S8, Obeng et al., 2022). However, both here and as previously reported, mitragynine alone did not significantly alter rectal temperature, suggesting that mitragynine in rats may be a low efficacy Aα2AR agonist as compared with clonidine and lofexidine, just as mitragynine is a low efficacy agonist at MOR. The differences in the in vitro and in vivo efficacy of mitragynine could be due to species differences (rats used in vivo versus human Aα2ARs used in vitro). While affinity and intrinsic activity of mitragynine have been reported for human and mouse MORs, no data are published from rat MORs, perhaps indicating a low level of interaction or response at the rat MORs. While the Aα2AR antagonists atipamezole and yohimbine at high doses (17.8 mg/kg) also produced hypothermia on their own, the main effect of these Aα2AR-selective ligands was to antagonize the effects of lofexidine and clonidine. Mitragynine, on the other hand, did not antagonize the effects of Aα2AR agonists but rather increased their potency, while exerting minimal effect on its own. This suggests that mitragynine binds to Aα2AR and is a low efficacy agonist in rats.
In contrast to the mitragynine-like discriminative stimulus effects, neither atipamezole nor naltrexone altered the rate-decreasing effects of mitragynine, indicating that the rate decreasing effects of mitragynine are not mediated by MOR or Aα2AR. Moreover, the doses of yohimbine and atipamezole that produced hypothermia and antinociception also produced significant decreases in response rates indicating that there may be a lack of pharmacological specificity (i.e., non-Aα2AR mediated hypothermia) at the higher doses (17.8 mg/kg) of atipamezole and yohimbine tested.
5. Conclusion
In summary, the present results suggest that mitragynine is not only a MOR agonist, but also an Aα2AR ligand with low efficacy agonist activity in vivo. The low efficacy MOR agonist (buprenorphine) is approved by the FDA to treat OUD, as is the Aα2R agonist lofexidine. Concurrent activation of either or both of these receptor types by mitragynine may contribute to the purported use of kratom and related products as a self-treatment for opioid withdrawal (Boyer et al., 2008). The potential dual activation of both MOR and Aα2AR by mitragynine may improve upon currently approved drugs for OUD by including both mechanisms in a single novel compound.
Supplementary Material
Acknowledgements
The authors would like to thank Samantha N. Hart at the College of Pharmacy, University of Florida, for administrative assistance.
Funding
This work was supported by National Institutes of Health National Institute on Drug Abuse [Grants DA25267, DA48353, and UG3/UH3 DA048353 01]; University of Florida Foundation; University of Florida Department of Pharmacodynamics Funding; and the Texas Tech University Health Sciences Center Office of Research and the Jerry H. Hodge School of Pharmacy. The views and opinions expressed in this manuscript are those of the authors only and do not necessarily represent the views, official policy or position of the U.S. Department of Health and Human Services or any of its affiliated institutions or agencies. Dr. Hampson was substantially involved with this work, consistent with his role as Scientific Officer of UG3 DA048353. He had no substantial involvement in other cited grants. TH was also partially supported by USPHS grant R01DA058018.
Footnotes
Ethics statement
This study was approved by the Institutional Animal Care and Use Committee (IACUC) at the University of Florida and was in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals, which was fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC).
CRediT authorship contribution statement
Samuel Obeng: Writing – original draft, Methodology, Investigation, Formal analysis, Data curation, Conceptualization. Morgan L. Crowley: Writing – review & editing, Resources. Marco Mottinelli: Writing – review & editing, Resources. Francisco León: Writing – review & editing, Resources. Julio D. Zuarth Gonzalez: Investigation, Formal analysis. Yiming Chen: Writing – review & editing, Data curation. Lea R. Gamez-Jimenez: Investigation. Luis F. Restrepo: Investigation, Formal analysis. Nicholas P. Ho: Investigation. Avi Patel: Investigation, Formal analysis. Joelma Martins Rocha: Investigation. Manuel A. Alvarez: Investigation. Amsha M. Thadisetti: Investigation. Chai R. Park: Investigation. Victoria L.C. Pallares: Investigation, Formal analysis. Megan J. Milner: Formal analysis. Clinton E. Canal: Writing – review & editing. Aidan J. Hampson: Writing – review & editing. Christopher R. McCurdy: Writing – review & editing, Resources, Funding acquisition, Conceptualization. Lance R. McMahon: Writing – review & editing, Supervision, Resources, Funding acquisition, Conceptualization. Jenny L. Wilkerson: Writing – review & editing, Supervision, Methodology, Conceptualization. Takato Hiranita: Writing – original draft, Visualization, Supervision, Methodology, Formal analysis, Data curation, Conceptualization.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.org/10.1016/j.ejphar.2024.176863.
Data availability
The data that support the findings of this study are available within the paper and its Supplemental Data. Any additional information is available on request from the corresponding authors.
References
- Boyer EW, Babu KM, Adkins JE, McCurdy CR, Halpern JH, 2008. Self-treatment of opioid withdrawal using kratom (Mitragynia speciosa korth). Addiction 103, 1048–1050. 10.1111/j.1360-0443.2008.02209.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bücheler MM, Hadamek K, Hein L, 2002. Two α2-adrenergic receptor subtypes, α2A and α2C, inhibit transmitter release in the brain of gene-targeted mice. Neuroscience 109, 819–826. 10.1016/S0306-4522(01)00531-0. [DOI] [PubMed] [Google Scholar]
- Butelman ER, Huang Y, Epstein DH, Shaham Y, Goldstein RZ, Volkow ND, Alia-Klein N, 2023. Overdose mortality rates for opioids and stimulant drugs are substantially higher in men than in women: state-level analysis. Neuropsychopharmacology 48, 1639–1647. 10.1038/s41386-023-01601-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chear NJ-Y, León F, Sharma A, Kanumuri SRR, Zwolinski G, Abboud KA, Singh D, Restrepo LF, Patel A, Hiranita T, Ramanathan S, Hampson AJ, McMahon LR, McCurdy CR, 2021. Exploring the chemistry of alkaloids from Malaysian Mitragyna speciosa (kratom) and the role of oxindoles on human opioid receptors. J. Nat. Prod 84, 1034–1043. 10.1021/acs.jnatprod.0c01055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng Y-C, Prusoff WH, 1973. Relationship between the inhibition constant (KI) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol 22, 3099–3108. 10.1016/0006-2952(73)90196-2. [DOI] [PubMed] [Google Scholar]
- Colpaert FC, 1978. Discriminative stimulus properties of narcotic analgesic drugs. Pharmacol. Biochem. Behav 9, 863–887. 10.1016/0091-3057(78)90370-2. [DOI] [PubMed] [Google Scholar]
- Costa T, Klinz F, Vachon L, Herz A, 1988. Opioid receptors are coupled tightly to G proteins but loosely to adenylate cyclase in NG108–15 cell membranes. Mol. Pharmacol 34, 744–754. [PubMed] [Google Scholar]
- Crimmins EM, Zhang YS, 2019. Aging populations, mortality, and life expectancy. Annu. Rev. Sociol 45, 69–89. 10.1146/annurev-soc-073117-041351. [DOI] [Google Scholar]
- Dang VC, Chieng BC, Christie MJ, 2012. Prolonged stimulation of μ-opioid receptors produces β-arrestin-2-mediated heterologous desensitization of α2-adrenoceptor function in locus ceruleus neurons. Mol. Pharmacol 82, 473–480. 10.1124/mol.112.079350. [DOI] [PubMed] [Google Scholar]
- Doughty B, Morgenson D, Brooks T, 2019. Lofexidine: a newly FDA-approved, nonopioid treatment for opioid withdrawal. Ann. Pharmacother 53, 746–753. 10.1177/10600280198289. [DOI] [PubMed] [Google Scholar]
- Drasner K, Fields HL, 1988. Synergy between the antinociceptive effects of intrathecal clonidine and systemic morphine in the rat. Pain 32, 309–312. 10.1016/0304-3959(88)90042-5. [DOI] [PubMed] [Google Scholar]
- Ellis CR, Racz R, Kruhlak NL, Kim MT, Zakharov AV, Southall N, Hawkins EG, Burkhart K, Strauss DG, Stavitskaya L, 2020. Evaluating kratom alkaloids using PHASE. PLoS One 15, e0229646. 10.1371/journal.pone.0229646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farkas DJ, Foss JD, Ward SJ, Rawls SM, 2022. Kratom alkaloid mitragynine: inhibition of chemotherapy-induced peripheral neuropathy in mice is dependent on sex and active adrenergic and opioid receptors. IBRO Neurosci. Rep 13, 198–206. 10.1016/j.ibneur.2022.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foss JD, Nayak SU, Tallarida CS, Farkas DJ, Ward SJ, Rawls SM, 2020. Mitragynine, bioactive alkaloid of kratom, reduces chemotherapy-induced neuropathic pain in rats through α-adrenoceptor mechanism. Drug Alcohol Depend. 209, 107946 10.1016/j.drugalcdep.2020.107946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Romeu A, Cox DJ, Smith KE, Dunn KE, Griffiths RR, 2020. Kratom (Mitragyna speciosa): user demographics, use patterns, and implications for the opioid epidemic. Drug Alcohol Depend. 208, 107849 10.1016/j.drugalcdep.2020.107849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gold MS, Redmond DE, Kleber HD, 1979. Noradrenergic hyperactivity in opiate withdrawal supported by clonidine reversal of opiate withdrawal. Am. J. Psychiatr 136, 100–102. 10.1176/ajp.136.1.100. [DOI] [PubMed] [Google Scholar]
- Harrison C, Traynor J, 2003. The [35S] GTPγS binding assay: approaches and applications in pharmacology. Life Sci. 74, 489–508. 10.1016/j.lfs.2003.07.005. [DOI] [PubMed] [Google Scholar]
- Harun N, Hassan Z, Navaratnam V, Mansor SM, Shoaib M, 2015. Discriminative stimulus properties of mitragynine (kratom) in rats. Psychopharmacology 232, 2227–2238. 10.1007/s00213-015-3866-5. [DOI] [PubMed] [Google Scholar]
- Hassan Z, Muzaimi M, Navaratnam V, Yusoff NH, Suhaimi FW, Vadivelu R, Vicknasingam BK, Amato D, von Hörsten S, Ismail NI, Jayabalan N, Hazim AI, Mansor SM, Müller CP, 2013. From Kratom to mitragynine and its derivatives: physiological and behavioural effects related to use, abuse, and addiction. Neurosci. Biobehav. Rev 37, 138–151. 10.1016/j.neubiorev.2012.11.012. [DOI] [PubMed] [Google Scholar]
- Hiranita T, Kohut SJ, Soto PL, Tanda G, Kopajtic TA, Katz JL, 2014. Preclinical efficacy of N-substituted benztropine analogs as antagonists of methamphetamine self-administration in rats. J. Pharmacol. Exp. Therapeut 348, 174–191. 10.1124/jpet.113.208264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hiranita T, Leon F, Felix JS, Restrepo LF, Reeves ME, Pennington AE, Obeng S, Avery BA, McCurdy CR, McMahon LR, Wilkerson JL, 2019. The effects of mitragynine and morphine on schedule-controlled responding and antinociception in rats. Psychopharmacology 236, 2725–2734. 10.1007/s00213-019-05247-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jalal H, Buchanich JM, Roberts MS, Balmert LC, Zhang K, Burke DS, 2018. Changing dynamics of the drug overdose epidemic in the United States from 1979 through 2016. Science 361, eaau1184. 10.1126/science.aau1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin Y, Elko E, Tran T, Yorio T, 1989. Inhibition of adenylate cyclase in bovine ciliary process and rabbit iris ciliary body to alpha2-adrenergic agonists. J. Ocul. Pharmacol. Therapeut 5, 189–197. 10.1089/jop.1989.5.189. [DOI] [PubMed] [Google Scholar]
- Jordan BA, Gomes I, Rios C, Filipovska J, Devi LA, 2003. Functional interactions between μ opioid and α2A-adrenergic receptors. Mol. Pharmacol 64, 1317–1324. 10.1124/mol.64.6.1317. [DOI] [PubMed] [Google Scholar]
- Kelly E, 2013. Ligand bias at the μ-opioid receptor. Biochem. Soc. Trans 41, 218–224. 10.1042/BST20120331. [DOI] [PubMed] [Google Scholar]
- Kruegel AC, Uprety R, Grinnell SG, Langreck C, Pekarskaya EA, Le Rouzic V, Ansonoff M, Gassaway MM, Pintar JE, Pasternak GW, Majumdar S, Sames D, 2019. 7-Hydroxymitragynine is an active metabolite of mitragynine and a key mediator of its analgesic effects. ACS Cent. Sci 5, 992–1001. 10.1021/acscentsci.9b00141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leff P, Dougall IG, 1993. Further concerns over Cheng-Prusoff analysis. Trends Pharmacol. Sci 14, 110–112. 10.1016/0165-6147(93)90080-4. [DOI] [PubMed] [Google Scholar]
- Macko E, Weisbach J, Douglas B, 1972. Some observations on the pharmacology of mitragynine. Arch. Int. Pharmacodyn. Ther 198, 145–161. [PubMed] [Google Scholar]
- Matsumoto K, Mizowaki M, Suchitra T, Murakami Y, Takayama H, Sakai S. i., Aimi N, Watanabe H, 1996a. Central antinociceptive effects of mitragynine in mice: contribution of descending noradrenergic and serotonergic systems. Eur. J. Pharmacol 317, 75–81. 10.1016/S0014-2999(96)00714-5. [DOI] [PubMed] [Google Scholar]
- Matsumoto K, Mizowaki M, Suchitra T, Takayama H, Sakai S. i., Aimi N, Watanabe H, 1996b. Antinociceptive action of mitragynine in mice: evidence for the involvement of supraspinal opioid receptors. Life Sci. 59, 1149–1155. 10.1016/0024-3205(96)00432-8. [DOI] [PubMed] [Google Scholar]
- Maxwell EA, King TI, Kamble SH, Raju KSR, Berthold EC, León F, Avery BA, McMahon LR, McCurdy CR, Sharma A, 2020. Pharmacokinetics and safety of mitragynine in beagle dogs. Planta Med. 86, 1278–1285. 10.1055/a-1212-5475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minor BG, Danysz W, Jonsson G, Mohammed AK, Post C, Archer T, 1989. Adaptive changes in α−2 adrenoceptor mediated responses: analgesia, hypothermia and hypoactivity. Pharmacol. Toxicol 65, 143–151. 10.1111/j.1600-0773.1989.tb01145.x. [DOI] [PubMed] [Google Scholar]
- Newton AC, Bootman MD, Scott JD, 2016. Second messengers. Cold Spring Harbor Perspect. Biol 8, a005926 10.1101/cshperspect.a005926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niedernberg A, Tunaru S, Blaukat A, Harris B, Kostenis E, 2003. Comparative analysis of functional assays for characterization of agonist ligands at G protein-coupled receptors. J. Biomol. Screen 8, 500–510. 10.1177/1087057103257555. [DOI] [PubMed] [Google Scholar]
- O’Rourke M, Blaxall H, Iversen L, Bylund D, 1994. Characterization of [3H] RX821002 binding to alpha-2 adrenergic receptor subtypes. J. Pharmacol. Exp. Therapeut 268, 1362–1367. [PubMed] [Google Scholar]
- Obeng S, Kamble SH, Reeves ME, Restrepo LF, Patel A, Behnke M, Chear NJY, Ramanathan S, Sharma A, León F, Hiranita T, Avery BA, McMahon LR, McCurdy CR, 2020. Investigation of the adrenergic and opioid binding affinities, metabolic stability, plasma protein binding properties, and functional effects of selected indole-based kratom alkaloids. J. Med. Chem 63, 433–439. 10.1021/acs.jmedchem.9b01465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obeng S, Leon F, Patel A, Gonzalez JDZ, Da Silva LC, Restrepo LF, Gamez-Jimenez LR, Ho NP, Calvache MPG, Pallares VL, Helmes JA, Shiomitsu SK, Soto PL, Hampson AJ, McCurdy CR, McMahon LR, Wilkerson JL, Hiranita T, 2022. Interactive effects of μ-opioid and adrenergic-α2 receptor agonists in rats: pharmacological investigation of the primary kratom alkaloid mitragynine and its metabolite 7-hydroxymitragynine. J. Pharmacol. Exp. Therapeut 383, 182–198. 10.1124/jpet.122.001192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obeng S, Wilkerson JL, León F, Reeves ME, Restrepo LF, Gamez-Jimenez LR, Patel A, Pennington AE, Taylor VA, Ho NP, Braun T, Fortner JD, Crowley ML, Williamson MR, Pallares VLC, Mottinelli M, Lopera-Londoño C, McCurdy CR, McMahon LR, Hiranita T, 2021. Pharmacological comparison of mitragynine and 7-hydroxymitragynine: in vitro affinity and efficacy for μ-opioid receptor and opioid-like behavioral effects in rats. J. Pharmacol. Exp. Therapeut 376, 410–427. 10.1124/jpet.120.000189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ossipov MH, Harris S, Lloyd P, Messineo E, 1990. An isobolographic analysis of the antinociceptive effect of systemically and intrathecally administered combinations of clonidine and opiates. J. Pharmacol. Exp. Therapeut 255, 1107–1116. [PubMed] [Google Scholar]
- Raffa RB, Pergolizzi JV, Taylor R, James RP, Pirner M, 2019. Differences in the receptor binding profile of lofexidine compared to clonidine. Pharmacol. Pharm 10, 1–10. 10.4236/pp.2019.101001. [DOI] [Google Scholar]
- Singh D, Narayanan S, Vicknasingam B, Prozialeck WC, Smith KE, Corazza O, Henningfield JE, Grundmann O, 2022. The use of kratom (Mitragyna speciosa Korth.) among people who co-use heroin and methamphetamine in Malaysia. J. Addiction Med 16, 223–228. 10.1097/ADM.0000000000000876. [DOI] [PubMed] [Google Scholar]
- Snedecor GW, Cochran WG, 1967. Statistical Methods, sixth ed. Iowa State University Press, Ames, Iowa. [Google Scholar]
- Stone LS, German JP, Kitto KF, Fairbanks CA, Wilcox GL, 2014. Morphine and clonidine combination therapy improves therapeutic window in mice: synergy in antinociceptive but not in sedative or cardiovascular effects. PLoS One 9, e109903. 10.1371/journal.pone.0109903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki T, Mori T, Tsuji M, Misawa M, Nagase H, 1995. Discriminative stimulus properties of morphine mediated by μ1-opioid receptors. Eur. J. Pharmacol 284, 195–198. 10.1016/0014-2999(95)00440-V. [DOI] [PubMed] [Google Scholar]
- Tal M, Silberstein A, Nusser E, 1985. Why does Coomassie Brilliant Blue R interact differently with different proteins? A partial answer. J. Biol. Chem 260, 9976–9980. 10.1016/S0021-9258(17)39198-6. [DOI] [PubMed] [Google Scholar]
- Tallarida RJ, 2002. The interaction index: a measure of drug synergism. Pain 98, 163–168. 10.1016/S0304-3959(02)00041-6. [DOI] [PubMed] [Google Scholar]
- Tan M, Walwyn WM, Evans CJ, Xie C-W, 2009. p38 MAPK and β-arrestin 2 mediate functional interactions between endogenous μ-opioid and α2A-adrenergic receptors in neurons. J. Biol. Chem 284, 6270–6281. 10.1074/jbc.M806742200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Timko C, Schultz NR, Cucciare MA, Vittorio L, Garrison-Diehn C, 2016. Retention in medication-assisted treatment for opiate dependence: a systematic review. J. Addict. Dis 35, 22–35. 10.1080/10550887.2016.1100960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ullman-Culleŕe MH, Foltz CJ, 1999. Body condition scoring: a rapid and accurate method for assessing health status in mice. Comp. Med 49, 319–323. [PubMed] [Google Scholar]
- Váradi A, Marrone GF, Palmer TC, Narayan A, Szabó MR, Le Rouzic V, Grinnell SG, Subrath JJ, Warner E, Kalra S, Hunkele A, Pagirsky J, Eans SO, Medina JM, Xu J, Pan Y-X, Borics A, Pasternak GW, McLaughlin JP, Majumdar S, 2016. Mitragynine/corynantheidine pseudoindoxyls as opioid analgesics with mu agonism and delta antagonism, which do not recruit β-arrestin-2. J. Med. Chem 59, 8381–8397. 10.1021/acs.jmedchem.6b00748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vicknasingam B, Narayanan S, Beng GT, Mansor SM, 2010. The informal use of ketum (Mitragyna speciosa) for opioid withdrawal in the northern states of peninsular Malaysia and implications for drug substitution therapy. Int. J. Drug Pol 21, 283–288. 10.1016/j.drugpo.2009.12.003. [DOI] [PubMed] [Google Scholar]
- Vilardaga J-P, Nikolaev VO, Lorenz K, Ferrandon S, Zhuang Z, Lohse MJ, 2008. Conformational cross-talk between α2A-adrenergic and μ-opioid receptors controls cell signaling. Nat. Chem. Biol 4, 126–131. 10.1038/nchembio.64. [DOI] [PubMed] [Google Scholar]
- Young AM, Masaki MA, Geula C, 1992. Discriminative stimulus effects of morphine: effects of training dose on agonist and antagonist effects of mu opioids. J. Pharmacol. Exp. Therapeut 261, 246–257. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data that support the findings of this study are available within the paper and its Supplemental Data. Any additional information is available on request from the corresponding authors.
