Mechanistic Understanding of Peptide Analogues, DALDA, [Dmt1]DALDA, and KGOP01, Binding to the Mu Opioid Receptor
The mu opioid receptor (MOR) is the primary target for analgesia of endogenous opioid peptides, alkaloids, synthetic small molecules with diverse scaffolds, and peptidomimetics. Peptide-based opioids are viewed as potential analgesics with reduced side effects and have received constant scientific interest over the years. This study focuses on three potent peptide and peptidomimetic MOR agonists, DALDA, [Dmt1]DALDA, and KGOP01, and the prototypical peptide MOR agonist DAMGO. We present the first molecular modeling study and structure–activity relationships aided by in vitro assays and molecular docking of the opioid peptide analogues, in order to gain insight into their mode of binding to the MOR. In vitro binding and functional assays revealed the same rank order with KGOP01 > [Dmt1]DALDA > DAMGO > DALDA for both binding and MOR activation. Using molecular docking at the MOR and three-dimensional interaction pattern analysis, we have rationalized the experimental outcomes and highlighted key amino acid residues responsible for agonist binding to the MOR. The Dmt (2′,6′-dimethyl-L-Tyr) moiety of [Dmt1]DALDA and KGOP01 was found to represent the driving force for their high potency and agonist activity at the MOR. These findings contribute to a deeper understanding of MOR function and flexible peptide ligand–MOR interactions, that are of significant relevance for the future design of opioid peptide-based analgesics.
Highlights
Opioids are the mainstay in the management of moderate to severe pain, and remain the most efficacious analgesics currently available [1]
Comparison of in Vitro Binding and Activation Profiles of DALDA, [Dmt ]DALDA, and KGOP01 to the approaches, we aimed to explain the molecular basis for their binding to the mu opioid receptor (MOR), in terms of understanding the structural correlations as well as interpreting the related
Given the essential clinical role of the MOR in mediating pain inhibition and other physiological activities, with endogenous peptides as natural agonists of the MOR, a basic understanding of the binding mechanism of opioid peptides to the MOR is required for their further development as potential analgesics and drugs for pain treatment and other human disorders
Summary
Opioids are the mainstay in the management of moderate to severe pain, and remain the most efficacious analgesics currently available [1]. Abuse and misuse of opioids became a significant public health concern due to the huge rise in overdose morbidity and mortality [6,7]. In this view, the development of effective and safer analgesics represents a key research goal for 21st century analgesic drug discovery and pain medicine
66
- 10.1016/s0014-2999(01)00946-3
- May 1, 2001
- European Journal of Pharmacology
1298
- 10.1038/nature10954
- Mar 21, 2012
- Nature
490
- 10.1002/prot.22234
- Jan 29, 2009
- Proteins: Structure, Function, and Bioinformatics
805
- 10.1038/s41594-017-0011-7
- Jan 1, 2018
- Nature Structural & Molecular Biology
72
- 10.1038/srep21548
- Feb 1, 2016
- Scientific Reports
10
- 10.1007/978-0-387-76678-2
- Jan 1, 2008
18
- 10.4155/fmc.14.132
- Dec 1, 2014
- Future Medicinal Chemistry
487
- 10.1001/jamapsychiatry.2018.3126
- Dec 5, 2018
- JAMA Psychiatry
361
- 10.1146/annurev-neuro-080317-061522
- May 31, 2018
- Annual Review of Neuroscience
675
- 10.1016/0014-2999(81)90364-2
- Apr 1, 1981
- European Journal of Pharmacology
- Research Article
61
- 10.1186/s12974-020-02029-3
- Nov 25, 2020
- Journal of Neuroinflammation
The existence of the neural control of mast cell functions has long been proposed. Mast cells (MCs) are localized in association with the peripheral nervous system (PNS) and the brain, where they are closely aligned, anatomically and functionally, with neurons and neuronal processes throughout the body. They express receptors for and are regulated by various neurotransmitters, neuropeptides, and other neuromodulators. Consequently, modulation provided by these neurotransmitters and neuromodulators allows neural control of MC functions and involvement in the pathogenesis of mast cell–related disease states. Recently, the roles of individual neurotransmitters and neuropeptides in regulating mast cell actions have been investigated extensively. This review offers a systematic review of recent advances in our understanding of the contributions of neurotransmitters and neuropeptides to mast cell activation and the pathological implications of this regulation on mast cell–related disease states, though the full extent to which such control influences health and disease is still unclear, and a complete understanding of the mechanisms underlying the control is lacking. Future validation of animal and in vitro models also is needed, which incorporates the integration of microenvironment-specific influences and the complex, multifaceted cross-talk between mast cells and various neural signals. Moreover, new biological agents directed against neurotransmitter receptors on mast cells that can be used for therapeutic intervention need to be more specific, which will reduce their ability to support inflammatory responses and enhance their potential roles in protecting against mast cell–related pathogenesis.
- Research Article
29
- 10.1038/s41598-020-70493-1
- Aug 14, 2020
- Scientific Reports
Pain remains a key therapeutic area with intensive efforts directed toward finding effective and safer analgesics in light of the ongoing opioid crisis. Amongst the neurotransmitter systems involved in pain perception and modulation, the mu-opioid receptor (MOR), a G protein-coupled receptor, represents one of the most important targets for achieving effective pain relief. Most clinically used opioid analgesics are agonists to the MOR, but they can also cause severe side effects. Medicinal plants represent important sources of new drug candidates, with morphine and its semisynthetic analogues as well-known examples as analgesic drugs. In this study, combining in silico (pharmacophore-based virtual screening and docking) and pharmacological (in vitro binding and functional assays, and behavioral tests) approaches, we report on the discovery of two naturally occurring plant alkaloids, corydine and corydaline, as new MOR agonists that produce antinociceptive effects in mice after subcutaneous administration via a MOR-dependent mechanism. Furthermore, corydine and corydaline were identified as G protein-biased agonists to the MOR without inducing β-arrestin2 recruitment upon receptor activation. Thus, these new scaffolds represent valuable starting points for future chemical optimization towards the development of novel opioid analgesics, which may exhibit improved therapeutic profiles.
- Research Article
13
- 10.3390/molecules26113267
- May 28, 2021
- Molecules
Opioids are the most effective analgesics, with most clinically available opioids being agonists to the µ-opioid receptor (MOR). The MOR is also responsible for their unwanted effects, including reward and opioid misuse leading to the current public health crisis. The imperative need for safer, non-addictive pain therapies drives the search for novel leads and new treatment strategies. In this study, the recently discovered MOR/nociceptin (NOP) receptor peptide hybrid KGNOP1 (H-Dmt-D-Arg-Aba-β-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) was evaluated following subcutaneous administration in mouse models of acute (formalin test) and chronic inflammatory pain (Complete Freund’s adjuvant-induced paw hyperalgesia), liabilities of spontaneous locomotion, conditioned place preference, and the withdrawal syndrome. KGNOP1 demonstrated dose-dependent antinociceptive effects in the formalin test, and efficacy in attenuating thermal hyperalgesia with prolonged duration of action. Antinociceptive effects of KGNOP1 were reversed by naltrexone and SB-612111, indicating the involvement of both MOR and NOP receptor agonism. In comparison with morphine, KGNOP1 was more potent and effective in mouse models of inflammatory pain. Unlike morphine, KGNOP1 displayed reduced detrimental liabilities, as no locomotor impairment nor rewarding and withdrawal effects were observed. Docking of KGNOP1 to the MOR and NOP receptors and subsequent 3D interaction pattern analyses provided valuable insights into its binding mode. The mixed MOR/NOP receptor peptide KGNOP1 holds promise in the effort to develop new analgesics for the treatment of various pain states with fewer MOR-mediated side effects, particularly abuse and dependence liabilities.
- Research Article
17
- 10.1021/acs.jmedchem.0c01915
- Jan 7, 2022
- Journal of Medicinal Chemistry
Strong opioid analgesics, including morphine, are the mainstays for treating moderate to severe acute pain and alleviating chronic cancer pain. However, opioid-related adverse effects, including nausea or vomiting, sedation, respiratory depression, constipation, pruritus (itch), analgesic tolerance, and addiction and abuse liability, are problematic. In addition, the use of opioids to relieve chronic noncancer pain is controversial due to the "opioid crisis" characterized by opioid misuse or abuse and escalating unintentional death rates due to respiratory depression. Hence, considerable research internationally has been aimed at the "Holy Grail" of the opioid analgesic field, namely the discovery of novel and safer opioid analgesics with improved opioid-related adverse effects. In this Perspective, medicinal chemistry strategies are addressed, where structurally diverse nonmorphinan-based opioid ligands derived from natural sources were deployed as lead molecules. The current state of play, clinical or experimental status, and novel opioid ligand discovery approaches are elaborated in the context of retaining analgesia with improved safety and reduced adverse effects, especially addiction liability.
- Supplementary Content
9
- 10.3390/ijms24087042
- Apr 11, 2023
- International Journal of Molecular Sciences
The United States is experiencing the most profound and devastating opioid crisis in history, with the number of deaths involving opioids, including prescription and illegal opioids, continuing to climb over the past two decades. This severe public health issue is difficult to combat as opioids remain a crucial treatment for pain, and at the same time, they are also highly addictive. Opioids act on the opioid receptor, which in turn activates its downstream signaling pathway that eventually leads to an analgesic effect. Among the four types of opioid receptors, the µ subtype is primarily responsible for the analgesic cascade. This review describes available 3D structures of the µ opioid receptor in the protein data bank and provides structural insights for the binding of agonists and antagonists to the receptor. Comparative analysis on the atomic details of the binding site in these structures was conducted and distinct binding interactions for agonists, partial agonists, and antagonists were observed. The findings in this article deepen our understanding of the ligand binding activity and shed some light on the development of novel opioid analgesics which may improve the risk benefit balance of existing opioids.
- Research Article
1
- 10.1002/chir.23619
- Sep 12, 2023
- Chirality
Even though chiral recognition for crown-ether CSPs is generally understood, on a molecular level, exact mechanisms for the resolution are still unclear. Furthermore, short peptide analytes often contain multiple amino moieties capable of binding to the crown ether selector. In order to extend the understanding in chiral recognition mechanisms, polar organic mode separation of Tyr-Arg-Phe-Lys-NH2 tetrapeptide llll/dddd enantiomers on S- and R-(3,3'-diphenyl-1,1'-binaphthyl)-20-crown-6 stationary phases was studied with 50-mM perchloric acid in methanol as mobile phase. Deviation from the generally acceptable 1:1 stoichiometry was supported by mass spectroscopy analysis of the formed complexes between tetrapeptide enantiomer and crown ether selectors, which revealed adducts possessing 1:1, 1:2, and 1:3 stoichiometry. Further investigation of complexation induced shifts by NMR indicated on different binding mechanisms between llll/dddd enantiomers of Tyr-Arg-Phe-Lys-NH2 and crown ether selectors. Enantioselective proton shifts were observed in studied tetrapeptide tyrosine and phenylalanine residues exclusively for llll enantiomer upon binding with S-(3,3'-diphenyl-1,1'-binaphthyl)-20-crown-6 selector (and dddd enantiomer with R-(3,3'-diphenyl-1,1'-binaphthyl)-20-crown-6 selector), indicating that these two amino acid residues contribute to chiral recognition. The obtained results were in agreement with the LC data.
- Research Article
9
- 10.3390/ijms21207738
- Oct 19, 2020
- International journal of molecular sciences
AA3266 is a hybrid compound consisting of opioid receptor agonist and neurokinin-1 receptor (NK1R) antagonist pharmacophores. It was designed with the desire to have an analgesic molecule with improved properties and auxiliary anticancer activity. Previously, the compound was found to exhibit high affinity for μ- and δ-opioid receptors, while moderate binding to NK1R. In the presented contribution, we report on a deeper investigation of this hybrid. In vivo, we have established that AA3266 has potent antinociceptive activity in acute pain model, comparable to that of morphine. Desirably, with prolonged administration, our hybrid induces less tolerance than morphine does. AA3266, contrary to morphine, does not cause development of constipation, which is one of the main undesirable effects of opioid use. In vitro, we have confirmed relatively strong cytotoxic activity on a few selected cancer cell lines, similar to or greater than that of a reference NK1R antagonist, aprepitant. Importantly, our compound affects normal cells to smaller extent what makes our compound more selective against cancer cells. In silico methods, including molecular docking, molecular dynamics simulations and fragment molecular orbital calculations, have been used to investigate the interactions of AA3266 with MOR and NK1R. Insights from these will guide structural optimization of opioid/antitachykinin hybrid compounds.
- Research Article
8
- 10.3390/molecules26175406
- Sep 6, 2021
- Molecules
Opioid agonists are well-established analgesics, widely prescribed for acute but also chronic pain. However, their efficiency comes with the price of drastically impacting side effects that are inherently linked to their prolonged use. To answer these liabilities, designed multiple ligands (DMLs) offer a promising strategy by co-targeting opioid and non-opioid signaling pathways involved in nociception. Despite being intimately linked to the Substance P (SP)/neurokinin 1 (NK1) system, which is broadly examined for pain treatment, the neurokinin receptors NK2 and NK3 have so far been neglected in such DMLs. Herein, a series of newly designed opioid agonist-NK2 or -NK3 antagonists is reported. A selection of reported peptidic, pseudo-peptidic, and non-peptide neurokinin NK2 and NK3 ligands were covalently linked to the peptidic μ-opioid selective pharmacophore Dmt-DALDA (H-Dmt-d-Arg-Phe-Lys-NH2) and the dual μ/δ opioid agonist H-Dmt-d-Arg-Aba-βAla-NH2 (KGOP01). Opioid binding assays unequivocally demonstrated that only hybrids SBL-OPNK-5, SBL-OPNK-7 and SBL-OPNK-9, bearing the KGOP01 scaffold, conserved nanomolar range μ-opioid receptor (MOR) affinity, and slightly reduced affinity for the δ-opioid receptor (DOR). Moreover, NK binding experiments proved that compounds SBL-OPNK-5, SBL-OPNK-7, and SBL-OPNK-9 exhibited (sub)nanomolar binding affinity for NK2 and NK3, opening promising opportunities for the design of next-generation opioid hybrids.
- Research Article
20
- 10.1021/acs.jmedchem.0c01376
- Sep 9, 2020
- Journal of Medicinal Chemistry
Fusionof nonopioid pharmacophores, such as neurotensin, with opioidligands represents an attractive approach for pain treatment. Herein,the μ-/δ-opioid agonist tetrapeptide H-Dmt-d-Arg-Aba-β-Ala-NH2 (KGOP01) was fused to NT(8-13) analogues.Since the NTS1 receptor has been linked to adverse effects, selectiveMOR-NTS2 ligands are preferred. Modifications were introduced withinthe native NT sequence, particularly a β3-homo aminoacid in position 8 and Tyr11 substitutions. Combinationof β3hArg and Dmt led to peptide 7,a MOR agonist, showing the highest NTS2 affinity described to date(Ki = 3 pM) and good NTS1 affinity (Ki = 4 nM), providing a >1300-fold NTS2 selectivity.The (6-OH)Tic-containing analogue 9 also exhibited highNTS2 affinity (Ki = 1.7 nM), with lowNTS1 affinity (Ki = 4.7 μM), resultingin an excellent NTS2 selectivity (>2700). In mice, hybrid 7 produced significant and prolonged antinociception (up to8 h),as compared to the KGOP01 opioid parent compound.
- Research Article
12
- 10.3390/biom12091241
- Sep 5, 2022
- Biomolecules
Despite various advantages, opioid peptides have been limited in their therapeutic uses due to the main drawbacks in metabolic stability, blood-brain barrier permeability, and bioavailability. Therefore, extensive studies have focused on overcoming the problems and optimizing the therapeutic potential. Currently, numerous peptide-based drugs are being marketed thanks to new synthetic strategies for optimizing metabolism and alternative routes of administration. This tutorial review briefly introduces the history and role of natural opioid peptides and highlights the key findings on their structure-activity relationships for the opioid receptors. It discusses details on opioid peptidomimetics applied to develop therapeutic candidates for the treatment of pain from the pharmacological and structural points of view. The main focus is the current status of various mimetic tools and the successful applications summarized in tables and figures.
- Research Article
44
- 10.1016/j.chest.2019.05.015
- Jun 10, 2019
- Chest
Solving the Opioid Crisis: Respiratory Depression by Opioids as Critical End Point
- Research Article
78
- 10.1074/jbc.m604278200
- Nov 1, 2006
- Journal of Biological Chemistry
In this study we investigated the mechanisms responsible for MAP kinase ERK1/2 activation following agonist activation of endogenous mu opioid receptors (MOR) normally expressed in cultured striatal neurons. Treatment with the MOR agonist fentanyl caused significant activation of ERK1/2 in neurons derived from wild type mice. Fentanyl effects were blocked by the opioid antagonist naloxone and were not evident in neurons derived from MOR knock-out (-/-) mice. In contrast, ERK1/2 activation by fentanyl was not evident in neurons from GRK3-/- mice or neurons pretreated with small inhibitory RNA for arrestin3. Consistent with this observation, treatment with the opiate morphine (which is less able to activate arrestin) did not elicit ERK1/2 activation in wild type neurons; however, transfection of arrestin3-(R170E) (a dominant positive form of arrestin that does not require receptor phosphorylation for activation) enabled morphine activation of ERK1/2. In addition, activation of ERK1/2 by fentanyl and morphine was rescued in GRK3-/- neurons following transfection with dominant positive arrestin3-(R170E). The activation of ERK1/2 appeared to be selective as p38 MAP kinase activation was not increased by either fentanyl or morphine treatment in neurons from wild type, MOR-/-, or GRK3-/- mice. In addition, U0126 (a selective inhibitor of MEK kinase responsible for ERK phosphorylation) blocked ERK1/2 activation by fentanyl. These results support the hypothesis that MOR activation of ERK1/2 requires opioid receptor phosphorylation by GRK3 and association of arrestin3 to initiate the cascade resulting in ERK1/2 phosphorylation in striatal neurons.
- Research Article
8
- 10.1097/aia.0000000000000268
- Jan 1, 2020
- International Anesthesiology Clinics
The role of opioids in cancer progression.
- Research Article
- 10.1016/j.pbb.2025.174056
- Sep 1, 2025
- Pharmacology, biochemistry, and behavior
KOR agonists for the treatment and/or prevention of opioid use disorder and cocaine use disorder.
- Research Article
74
- 10.1038/sj.npp.1301675
- Jan 23, 2008
- Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology
Amygdala opioids such as enkephalin appear to play some role in the control of anxiety and the anxiolytic effects of benzodiazepines, although the opioid receptor subtypes mediating such effects are unclear. This study compared the influences of mu-opioid receptor (MOR) activation in the central nucleus of the amygdala (CEA) on unconditioned fear or anxiety-like responses in two models, the elevated plus maze, and the defensive burying test. The role of MORs in the anxiolytic actions of the benzodiazepine agonist diazepam was also examined using both models. Either the MOR agonist [D-Ala(2), NMe-Phe(4), Gly-ol(5)]-enkephalin (DAMGO), or the MOR antagonists Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP) or beta-funaltrexamine (FNA) were bilaterally infused into the CEA of rats before testing. The results show that microinjection of DAMGO in the CEA decreased open-arm time in the plus maze, whereas CTAP increased open-arm behaviors. In contrast, DAMGO injections in the CEA reduced burying behaviors and increased rearing following exposure to a predator odor, suggesting a shift in the behavioral response in this context. Amygdala injections of the MOR agonist DAMGO or the MOR antagonist CTAP failed to change the anxiolytic effects of diazepam in either test. Our results demonstrate that MOR activation in the central amygdala exerts distinctive effects in two different models of unconditioned fear or anxiety-like responses, and suggest that opioids may exert context-specific regulation of amygdalar output circuits and behavioral responses during exposure to potential threats (open arms of the maze) vs discrete threats (predator odor).
- Research Article
30
- 10.1111/jnc.12976
- Nov 17, 2014
- Journal of Neurochemistry
Physical exercise stimulates the release of endogenous opioid peptides supposed to be responsible for changes in mood, anxiety, and performance. Exercise alters sensitivity to these effects that modify the efficacy at the opioid receptor. Although there is evidence that relates exercise to neuropeptide expression in the brain, the effects of exercise on opioid receptor binding and signal transduction mechanisms downstream of these receptors have not been explored. Here, we characterized the binding and G protein activation of mu opioid receptor, kappa opioid receptor or delta opioid receptor in several brain regions following acute (7 days) and chronic (30 days) exercise. As regards short- (acute) or long-term effects (chronic) of exercise, overall, higher opioid receptor binding was observed in acute-exercise animals and the opposite was found in the chronic-exercise animals. The binding of [(35) S]GTPγS under basal conditions (absence of agonists) was elevated in sensorimotor cortex and hippocampus, an effect more evident after chronic exercise. Divergence of findings was observed for mu opioid receptor, kappa opioid receptor, and delta opioid receptor receptor activation in our study. Our results support existing evidence of opioid receptor binding and G protein activation occurring differentially in brain regions in response to diverse exercise stimuli. We characterized the binding and G protein activation of mu, kappa, and delta opioid receptors in several brain regions following acute (7 days) and chronic (30 days) exercise. Higher opioid receptor binding was observed in the acute exercise animal group and opposite findings in the chronic exercise group. Higher G protein activation under basal conditions was noted in rats submitted to chronic exercise, as visible in the depicted pseudo-color autoradiograms.
- Research Article
- 10.1096/fasebj.2020.34.s1.05714
- Apr 1, 2020
- The FASEB Journal
Pharmacological modulation of the delta opioid receptor (DOR) in conjunction with a mu‐opioid receptor (MOR) agonist produces antinociceptive effects with potentially a better side effect profile. MMP2200 (a.k.a.Lactomorphin) is a CNS permeable, glycosylated peptide and an agonist at both MORs and DORs. Previous reports characterized the MOR agonist effects of MMP2200; however, the DOR agonist effects of MMP2200in vivo have not been explored. DOR agonists, such as SNC80, have been shown to produce convulsions and discriminative stimulus effects in rodents. Therefore, based on the DOR agonist activity of MMP2200, we hypothesized that MMP2200 would produce DOR‐mediated convulsions in mice and SNC80‐like discriminative stimulus effects in rats. Convulsions were evaluated by continual observation for up to 60 min in an observation cage. For SNC80 discrimination experiments, rats were trained to discriminate an injection of 3.2 mg/kg SNC80 from saline under a fixed ratio 10 schedule of food reinforcement, and different doses of SNC80 or MMP2200 were substituted to evaluate percent generalization and response rates. MMP2200 produced brief, non‐lethal convulsions in C57BL/6N, but not ICR, mice that were blocked by the DOR antagonist naltrindole and by genetic deletion of DOR, indicating that MMP2200‐induced convulsions are DOR mediated. Following acute drug administration, mice were then given a second dose of the DOR agonist 24 h later. These mice were tolerant to convulsions induced by SNC80, but not to MMP2200‐induced convulsions. Tolerance to the convulsive effects of MMP2200 was apparent 8 h following the initial injection. SNC80 induced convulsions were enhanced in mice lacking arrestin 2 but were unaltered in mice lacking arrestin 3. However, MMP2200‐induced convulsions were attenuated in mice lacking either arrestin 2 or 3. In rats trained to discriminate SNC80, MMP2200 fully generalized to the discriminative stimulus effects of SNC80 and only slightly decreased rates of responding. Overall, these results demonstrate that the dual MOR/DOR agonist MMP2200 produces many behavioral effects that are similar to the prototypical DOR agonist SNC80. Interestingly, there are some distinct differences in these DOR agonist effects, which may be mediated by concurrent MOR agonist activity with MMP2200 or by activating differential intracellular signaling molecules downstream of DOR activation. Future studies will evaluate the effects of DOR agonists and signaling pathways mediating these effects.
- Research Article
33
- 10.1021/acschemneuro.8b00349
- Aug 29, 2018
- ACS Chemical Neuroscience
Structure-activity relationship (SAR) studies of numerous opioid ligands have shown that introduction of a methyl or ethyl group on the tertiary amino group at position 17 of the epoxymorphinan skeleton generally results in a mu opioid receptor (MOR) agonist while introduction of a cyclopropylmethyl group typically leads to an antagonist. Furthermore, it has been shown that introduction of heterocyclic ring systems at position 6 can favor antagonism. However, it was reported that 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(2'-indolyl)acetamido]morphinan (INTA), which bears a cyclopropylmethyl group at position 17 and an indole ring at position 6, acted as a MOR agonist. We herein report a SAR study on INTA with a series of its complementary derivatives to understand how introduction of an indole moiety with α or β linkage at position 6 of the epoxymorphinan skeleton may influence ligand function. Interestingly, one of INTA derivatives, compound 15 (NAN) was identified as a MOR antagonist both in vitro and in vivo. Molecular modeling studies revealed that INTA and NAN may interact with different domains of the MOR allosteric binding site. In addition, INTA may interact with W293 and N150 residues found in the orthosteric site to stabilize MOR activation conformation while NAN does not. These results suggest that INTA and NAN may be bitopic ligands and the type of allosteric interactions with the MOR influence their functional activity. These insights along with our enriched comprehension of the "message-address" concept will to benefit future ligand design.
- Research Article
14
- 10.1016/j.neuint.2019.104521
- Aug 13, 2019
- Neurochemistry International
Activation of MORs in the VTA induces changes on cFos expression in different projecting regions: Effect of inflammatory pain
- Research Article
- 10.1038/s41386-025-02214-z
- Sep 3, 2025
- Neuropsychopharmacology
The development of safer mu opioid receptor (MOR) agonists with reduced side effects is a key focus of pain research. Some studies have suggested that MOR agonists with reduced β-arrestin 2 (βArr2) signaling (i.e. G-protein biased agonists) may have greater therapeutic windows. However, there have been a several conflicting reports, and it is not clear what role, if any, βArr2 signaling plays in MOR-mediated analgesia, tolerance, or side effects. Therefore, we used βArr2 knockout mice to systematically investigate the causal role of βArr2 signaling in antinociception, antinociceptive tolerance, respiratory depression, constipation, and reward induced by morphine and the two novel MOR agonists, kurkinorin and kurkinol. Kurkinorin and kurkinol exhibited potent antinociceptive effects that were reversed by MOR knockout. Unlike morphine or kurkinorin, our most G-protein biased agonist, kurkinol, showed no significant tolerance after seven days of ~2×ED50 dosing. However, in a chemotherapy-induced neuropathic pain model, all three compounds were ineffective after 20 days of ~ED50 dosing, indicative of tolerance. All compounds exhibited significant MOR-dependent side effects, though kurkinorin had reduced gastrointestinal and respiratory depressive effects compared to morphine despite exhibiting less G-protein bias. Knockout of βArr2 significantly increased antinociceptive potency for morphine and kurkinorin but not kurkinol, and otherwise had no significant impact on tolerance or any side effect tested. These results largely suggest that βArr2 signaling does not drive MOR-mediated antinociceptive tolerance, respiratory depression, constipation, or reward and do not support the development of G-protein biased compounds as a broadly effective strategy to reduce side effects.
- Research Article
4
- 10.3389/fpsyt.2023.1186397
- May 23, 2023
- Frontiers in Psychiatry
IntroductionTianeptine is approved in some countries to treat depression and anxiety. In addition to its activity on serotonin and glutamate neurotransmission, tianeptine has been proven to be a mu-opioid receptor (MOR) agonist, but only a few preclinical studies have characterized the opioid-like behavioral effects of tianeptine.MethodsIn this study, we tested tianeptine activity on G protein activation using the [S35] GTPγS binding assay in brain tissue from MOR+/+ and MOR−/− mice. Then, to determine whether tianeptine behavioral responses are MOR-dependent, we characterized the analgesic, locomotor, and rewarding responses of tianeptine in MOR+/+ and MOR−/− mice using tail immersion, hot plate, locomotor, and conditioned place preference tests.ResultsUsing the [S35] GTPγS binding assay, we found that tianeptine signaling is mediated by MOR in the brain with properties similar to those of DAMGO (a classic MOR agonist). Furthermore, we found that the MOR is necessary for tianeptine's analgesic (tail immersion and hot plate), locomotor, and rewarding (conditioned place preference) effects. Indeed, these behavioral effects could only be measured in MOR+/+ mice but not in MOR−/− mice. Additionally, chronic administration of tianeptine induced tolerance to its analgesic and hyperlocomotor effects.DiscussionThese findings suggest that tianeptine's opioid-like effects require MOR and that chronic use could lead to tolerance.
- Research Article
5
- 10.1016/j.pscychresns.2022.111505
- Jun 6, 2022
- Psychiatry research. Neuroimaging
Rejection sensitivity and mu opioid receptor dynamics associated with mood alterations in response to social feedback
- Research Article
186
- 10.1371/journal.pone.0042365
- Jul 31, 2012
- PLoS ONE
The canonical two neuron model of opioid reward posits that mu opioid receptor (MOR) activation produces reward by disinhibiting midbrain ventral tegmental area (VTA) dopamine neurons through inhibition of local GABAergic interneurons. Although indirect evidence supports the neural circuit postulated by this model, its validity has been called into question by growing evidence for VTA neuronal heterogeneity and the recent demonstration that MOR agonists inhibit GABAergic terminals in the VTA arising from extrinsic neurons. In addition, VTA MOR reward can be dopamine-independent. To directly test the assumption that MOR activation directly inhibits local GABAergic neurons, we investigated the properties of rat VTA GABA neurons directly identified with either immunocytochemistry for GABA or GAD65/67, or in situ hybridization for GAD65/67 mRNA. Utilizing co-labeling with an antibody for the neural marker NeuN and in situ hybridization against GAD65/67, we found that 23±3% of VTA neurons are GAD65/67(+). In contrast to the assumptions of the two neuron model, VTA GABAergic neurons are heterogeneous, both physiologically and pharmacologically. Importantly, only 7/13 confirmed VTA GABA neurons were inhibited by the MOR selective agonist DAMGO. Interestingly, all confirmed VTA GABA neurons were insensitive to the GABAB receptor agonist baclofen (0/6 inhibited), while all confirmed dopamine neurons were inhibited (19/19). The heterogeneity of opioid responses we found in VTA GABAergic neurons, and the fact that GABA terminals arising from neurons outside the VTA are inhibited by MOR agonists, make further studies essential to determine the local circuit mechanisms underlying VTA MOR reward.
- Research Article
27
- 10.1007/s00213-005-0030-7
- Jun 29, 2005
- Psychopharmacology
Heroin is rapidly metabolized to morphine that in turn is transformed in morphine-3-glucuronide (M3G), an inactive metabolite, and morphine-6-glucuronide (M6G), a potent mu-opioid receptor (MOR) agonist. We have found that heroin addicts exhibit higher M6G/M3G ratios relative to morphine-treated control subjects. We have also shown that heroin-treated rats exhibit measurable levels of M6G (which is usually undetectable in this species) and reduced levels of M3G. We investigated the role of MOR in these effects of heroin, by examining the effects of methadone, a MOR agonist, and of naltrexone, a MOR antagonist, on morphine glucuronidation. We also investigated the effects of alcohol, which is known to alter drug metabolism and is frequently coabused by heroin addicts. Morphine glucuronidation was studied in liver microsomes obtained from rats exposed daily for 10 days to saline, heroin (10 mg/kg, i.p.), naltrexone (20-40 mg/kg, i.p.), heroin + naltrexone (10 mg/kg+20-40 mg/kg, i.p.), methadone (5-20 mg/kg, i.p.), or 10% ethanol. Heroin induced the synthesis of M6G and decreased the synthesis of M3G. Naltrexone exhibited intrinsic modulatory activity on morphine glucuronidation, increasing the synthesis of M3G via a low-affinity/high-capacity reaction characterized by positive cooperativity. The rate of M3G synthesis in the heroin + naltrexone groups was not different from that of the naltrexone groups. Methadone and ethanol induced a modest increase in M3G synthesis and had no effect on M6G synthesis. The effects of heroin on morphine glucuronidation are not shared by methadone or alcohol (two drugs that figure prominently in the natural history of heroin addiction) and do not appear to depend on the activation of MOR.
- Research Article
12
- 10.1016/j.brainres.2012.10.024
- Oct 23, 2012
- Brain Research
Herkinorin dilates cerebral vessels via kappa opioid receptor and cyclic adenosine monophosphate (cAMP) in a piglet model
- New
- Research Article
- 10.3390/molecules30214313
- Nov 6, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214314
- Nov 6, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214311
- Nov 6, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214320
- Nov 6, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214298
- Nov 5, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214293
- Nov 5, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214307
- Nov 5, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214292
- Nov 5, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214297
- Nov 5, 2025
- Molecules
- New
- Research Article
- 10.3390/molecules30214310
- Nov 5, 2025
- Molecules
- Ask R Discovery
- Chat PDF