Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.804
Filtrer
1.
Molecules ; 29(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38998913

RÉSUMÉ

This study explored the potential of a series of PZM21 analogues for pain treatment. Specifically, the hydroxyphenyl ring of PZM21 was replaced with a naphthyl ring, the thienyl ring was substituted with either a phenyl ring or furan rings, and the essential dimethylamine and urea groups were retained. These compounds aimed to enhance safety and minimize the adverse effects associated with opioid drugs. The research findings suggest that compound 6a does not induce ß-arrestin recruitment at low-nanomolar concentrations but exhibits significant analgesic effects in established mouse models. Compared to morphine, 6a shows advantages in alleviating respiratory depression and minimizing physical dependence. Molecular docking studies underscore the pivotal role of the D147 amino acid residue in the analgesic mechanism of 6a. Consequently, 6a is a compelling candidate for the development of safer opioid analgesics and warrants further attention.


Sujet(s)
Analgésiques morphiniques , Simulation de docking moléculaire , Récepteur mu , Récepteur mu/agonistes , Récepteur mu/métabolisme , Animaux , Souris , Analgésiques morphiniques/pharmacologie , Analgésiques morphiniques/composition chimique , Analgésiques morphiniques/synthèse chimique , Humains , Relation structure-activité , Douleur/traitement médicamenteux , Mâle , Structure moléculaire , Thiophènes , Urée/analogues et dérivés
2.
Sci Rep ; 14(1): 16250, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009645

RÉSUMÉ

[11C]Carfentanil ([11C]CFN) is the only selective carbon-11 labeled radiotracer currently available for positron emission tomography (PET) imaging of mu opioid receptors (MORs). Though used extensively in clinical research, [11C]CFN has not been thoroughly characterized as a tool for preclinical PET imaging. As we were occasionally observing severe vital sign instability in rat [11C]CFN studies, we set out to investigate physiological effects of CFN mass and to explore its influence on MOR quantification. In anesthetized rats (n = 15), significant dose-dependent PCO2 increases and heart rate decreases were observed at a conventional tracer dose range (IV, > 100 ng/kg). Next, we conducted baseline and retest [11C]CFN PET scans over a wide range of molar activities. Baseline [11C]CFN PET studies (n = 27) found that nondisplaceable binding potential (BPND) in the thalamus was positively correlated to CFN injected mass, demonstrating increase of MOR availability at higher injected CFN mass. Consistently, when CFN injected mass was constrained < 40 ng/kg (~ 10% MOR occupancy in rats), baseline MOR availability was significantly decreased. For test-retest variability (TRTV), better reproducibility was achieved by controlling CFN injected mass to limit the difference between scans. Taken together, we report significant cardiorespiratory depression and a paradoxical influence on baseline MOR availability at conventional tracer doses in rats. Our findings might reflect changes in cerebral blood flow, changes in receptor affinity, or receptor internalization, and merits further mechanistic investigation. In conclusion, rat [11C]CFN PET requires stringent quality assurance of radiotracer synthesis and mass injected to avoid pharmacological effects and limit potential influences on MOR quantification and reproducibility.


Sujet(s)
Encéphale , Radio-isotopes du carbone , Fentanyl , Tomographie par émission de positons , Récepteur mu , Animaux , Récepteur mu/métabolisme , Fentanyl/analogues et dérivés , Fentanyl/métabolisme , Fentanyl/pharmacologie , Rats , Tomographie par émission de positons/méthodes , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Mâle , Rat Sprague-Dawley , Radiopharmaceutiques/pharmacocinétique
3.
Exp Dermatol ; 33(6): e15096, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38922774

RÉSUMÉ

While the evidence for the implication of opioid receptors (OPr) in ageing is growing, there is, to our knowledge, no study focusing directly on changes in vivo cutaneous OPr expression with increasing age. We thus investigated OPr expression in 30 healthy female Asian volunteers in Southern China whose ages range from the early 20s to the early 60s. Excisional biopsies were taken from the sun-exposed extensor area of the lower arm and the photo-protected area of the upper inner arm. The thickness of the epidermal layers, melanin content, as well as expression of mu-opioid receptors (MOPr) and delta-opioid receptors (DOPr) were compared between different age ranges and photo-exposure status. Significant increased epidermal hypertrophy on the extensor surface was observed. There was significant reduction of DOPr in the epidermis with increasing age, independent of photo-ageing. The increase of melanin was significantly correlated with epidermal DOPr expression, not with MOPr expression. DOPr expression could thus serve as a marker for real biological ageing unaffected by chronic photo-exposure. Additionally, DOPr expression was inversely correlated with the deposition of melanin. Based on these results, we hypothesise that regulation of DOPr expression could be used to improve aged skin, including hyperpigmentation.


Sujet(s)
Asiatiques , Mélanines , Récepteur delta , Vieillissement de la peau , Humains , Femelle , Mélanines/métabolisme , Mélanines/biosynthèse , Adulte , Récepteur delta/métabolisme , Adulte d'âge moyen , Jeune adulte , Épiderme/métabolisme , Récepteur mu/métabolisme , Chine
4.
Mol Pain ; 20: 17448069241260348, 2024.
Article de Anglais | MEDLINE | ID: mdl-38828868

RÉSUMÉ

Hyperalgesic priming is a preclinical model of the transition from acute to chronic pain characterized by a leftward shift in the dose-response curve for and marked prolongation of prostaglandin E2 (PGE2)-induced mechanical hyperalgesia, in vivo. In vitro, priming in nociceptors is characterized by a leftward shift in the concentration dependence for PGE2-induced nociceptor sensitization. In the present in vitro study we tested the hypothesis that a mu-opioid receptor (MOR) agonist opioid analgesic, morphine, can produce priming by its direct action on nociceptors. We report that treatment of nociceptors with morphine, in vitro, produces a leftward shift in the concentration dependence for PGE2-induced nociceptor sensitization. Our findings support the suggestion that opioids act directly on nociceptors to induce priming.


Sujet(s)
Dinoprostone , Morphine , Nocicepteurs , Morphine/pharmacologie , Animaux , Nocicepteurs/effets des médicaments et des substances chimiques , Nocicepteurs/métabolisme , Dinoprostone/métabolisme , Dinoprostone/pharmacologie , Récepteur mu/métabolisme , Analgésiques morphiniques/pharmacologie , Mâle , Rats , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Ganglions sensitifs des nerfs spinaux/métabolisme , Hyperalgésie/induit chimiquement , Hyperalgésie/traitement médicamenteux , Rat Sprague-Dawley , Relation dose-effet des médicaments
5.
Adv Neurobiol ; 35: 221-239, 2024.
Article de Anglais | MEDLINE | ID: mdl-38874725

RÉSUMÉ

Physical exercise is often cited as an important part of an intervention for depression, and there is empirical evidence to support this. However, the mechanism of action through which any potential antidepressant effects are produced is not widely understood. Recent evidence points toward the involvement of endogenous opioids, and especially the mu-opioid system, as a partial mediator of these effects. In this chapter, we discuss the current level of empirical support for physical exercise as either an adjunctive or standalone intervention for depression. We then review the extant evidence for involvement of endogenous opioids in the proposed antidepressant effects of exercise, with a focus specifically on evidence for mu-opioid system involvement.


Sujet(s)
Traitement par les exercices physiques , Exercice physique , Récepteur mu , Humains , Récepteur mu/métabolisme , Exercice physique/physiologie , Traitement par les exercices physiques/méthodes , Dépression/thérapie , Dépression/métabolisme , Résultat thérapeutique
6.
Adv Neurobiol ; 35: 287-313, 2024.
Article de Anglais | MEDLINE | ID: mdl-38874729

RÉSUMÉ

Kratom (Mitragyna speciosa) is a substance derived from botanical compounds native to Southeast Asia. This substance has been cultivated predominantly in Thailand, Malaysia, Vietnam, and Myanmar, where it has historically been used in traditional medicine as a near panacea for several health problems. Such ritualistic use of kratom has been present for centuries; however, recreational use appears to have increased globally, especially in the United States. Pharmacodynamic and pharmacokinetic studies have found that kratom demonstrates a unique parabolic, dose-dependent pattern of effects ranging from stimulation to opioid and analgesic effects. Pharmacological research indicates that kratom is both a mu opioid receptor (µ-OR; MOR) and a kappa opioid receptor (κ-OR; KOR) agonist, which mediates its analgesic effects. Other research suggests that kratom may simultaneously act on dopaminergic and serotonergic receptors, which mediate its stimulant effects. This chapter reviews the literature related to the structural, functional, and cultural characteristics of kratom use. We begin with an overview of current and historical patterns of kratom, followed by a review of data on the pharmacodynamics and pharmacokinetics of kratom thus far.


Sujet(s)
Mitragyna , Extraits de plantes , Récepteur kappa , Humains , Extraits de plantes/pharmacologie , Animaux , Récepteur kappa/métabolisme , Récepteur kappa/agonistes , Analgésiques morphiniques/pharmacologie , Analgésiques morphiniques/usage thérapeutique , Récepteur mu/métabolisme , Récepteur mu/agonistes , Asie du Sud-Est
7.
Genes Brain Behav ; 23(3): e12906, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38861664

RÉSUMÉ

Motherhood is a costly life-history transition accompanied by behavioral and neural plasticity necessary for offspring care. Motherhood in the monogamous prairie vole is associated with decreased pair bond strength, suggesting a trade-off between parental investment and pair bond maintenance. Neural mechanisms governing pair bonds and maternal bonds overlap, creating possible competition between the two. We measured mRNA expression of genes encoding receptors for oxytocin (oxtr), dopamine (d1r and d2r), mu-opioids (oprm1a), and kappa-opioids (oprk1a) within three brain areas processing salience of sociosensory cues (anterior cingulate cortex; ACC), pair bonding (nucleus accumbens; NAc), and maternal care (medial preoptic area; MPOA). We compared gene expression differences between pair bonded prairie voles that were never pregnant, pregnant (~day 16 of pregnancy), and recent mothers (day 3 of lactation). We found greater gene expression in the NAc (oxtr, d2r, oprm1a, and oprk1a) and MPOA (oxtr, d1r, d2r, oprm1a, and oprk1a) following the transition to motherhood. Expression for all five genes in the ACC was greatest for females that had been bonded for longer. Gene expression within each region was highly correlated, indicating that oxytocin, dopamine, and opioids comprise a complimentary gene network for social signaling. ACC-NAc gene expression correlations indicated that being a mother (oxtr and d1r) or maintaining long-term pair bonds (oprm1a) relies on the coordination of different signaling systems within the same circuit. Our study suggests the maternal brain undergoes changes that prepare females to face the trade-off associated with increased emotional investment in offspring, while also maintaining a pair bond.


Sujet(s)
Arvicolinae , Comportement maternel , Noyau accumbens , Monogamie , Récepteur mu , Animaux , Femelle , Arvicolinae/génétique , Récepteur mu/génétique , Récepteur mu/métabolisme , Comportement maternel/physiologie , Noyau accumbens/métabolisme , Grossesse , Récepteurs à l'ocytocine/génétique , Récepteurs à l'ocytocine/métabolisme , Récepteur kappa/génétique , Récepteur kappa/métabolisme , Gyrus du cingulum/métabolisme , Aire préoptique/métabolisme , Récepteur dopamine D1/génétique , Récepteur dopamine D1/métabolisme
8.
Science ; 384(6700): eadn0886, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843332

RÉSUMÉ

In addition to their intrinsic rewarding properties, opioids can also evoke aversive reactions that protect against misuse. Cellular mechanisms that govern the interplay between opioid reward and aversion are poorly understood. We used whole-brain activity mapping in mice to show that neurons in the dorsal peduncular nucleus (DPn) are highly responsive to the opioid oxycodone. Connectomic profiling revealed that DPn neurons innervate the parabrachial nucleus (PBn). Spatial and single-nuclei transcriptomics resolved a population of PBn-projecting pyramidal neurons in the DPn that express µ-opioid receptors (µORs). Disrupting µOR signaling in the DPn switched oxycodone from rewarding to aversive and exacerbated the severity of opioid withdrawal. These findings identify the DPn as a key substrate for the abuse liability of opioids.


Sujet(s)
Analgésiques morphiniques , Apprentissage par évitement , Troubles liés aux opiacés , Oxycodone , Noyau parabrachial , Cortex préfrontal , Récepteur mu , Récompense , Animaux , Mâle , Souris , Analgésiques morphiniques/pharmacologie , Connectome , Souris de lignée C57BL , Neurones/métabolisme , Neurones/physiologie , Troubles liés aux opiacés/métabolisme , Oxycodone/pharmacologie , Noyau parabrachial/métabolisme , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/physiologie , Cellules pyramidales/métabolisme , Récepteur mu/métabolisme , Récepteur mu/génétique , Syndrome de sevrage/métabolisme , Transcriptome
9.
J Med Chem ; 67(12): 10447-10463, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38869493

RÉSUMÉ

In recent years, synthetic opioids have emerged as a predominant cause of drug-overdose-related fatalities, causing the "opioid crisis." To design safer therapeutic agents, we accidentally discovered µ-opioid receptor (MOR) antagonists based on fentanyl with a relatively uncomplicated chemical composition that potentiates structural modifications. Here, we showed the development of novel atropisomeric fentanyl analogues that exhibit more potent antagonistic activity against MOR than naloxone, a morphinan MOR antagonist. Derivatives displaying stable axial chirality were synthesized based on the amide structure of fentanyl. The aS- and aR-enantiomers exerted antagonistic and agonistic effects on the MOR, respectively, and each atropisomer interacted with the MOR by assuming a distinct binding mode through molecular docking. These findings suggest that introducing atropisomerism into fentanyl may serve as a key feature in the molecular design of future MOR antagonists to help mitigate the opioid crisis.


Sujet(s)
Fentanyl , Récepteur mu , Récepteur mu/antagonistes et inhibiteurs , Récepteur mu/métabolisme , Fentanyl/pharmacologie , Fentanyl/analogues et dérivés , Fentanyl/composition chimique , Stéréoisomérie , Humains , Simulation de docking moléculaire , Relation structure-activité , Animaux , Antagonistes narcotiques/composition chimique , Antagonistes narcotiques/pharmacologie , Conformation moléculaire , Analgésiques morphiniques/pharmacologie , Analgésiques morphiniques/composition chimique , Analgésiques morphiniques/synthèse chimique , Cellules CHO , Cricetulus
11.
Nat Commun ; 15(1): 5372, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918367

RÉSUMÉ

The synthesis of constrained 12-membered rings is notably difficult. The main challenges result from constraints during the linear peptide cyclization. Attempts to overcome constraints through excessive activation frequently cause peptidyl epimerization, while insufficient activation of the C-terminus hampers cyclization and promotes intermolecular oligomer formation. We present a ß-thiolactone framework that enables the synthesis of cyclo-tetrapeptides via direct aminolysis. This tactic utilizes a mechanism that restricts C-terminal carbonyl rotation while maintaining high reactivity, thereby enabling efficient head-to-tail amidation, reducing oligomerization, and preventing epimerization. A broad range of challenging cyclo-tetrapeptides ( > 20 examples) are synthesized in buffer and exhibits excellent tolerance toward nearly all proteinogenic amino acids. Previously unattainable macrocycles, such as cyclo-L-(Pro-Tyr-Pro-Val), have been produced and identified as µ-opioid receptor (MOR) agonists, with an EC50 value of 2.5 nM. Non-epimerizable direct aminolysis offers a practical solution for constrained peptide cyclization, and the discovery of MOR agonist activity highlights the importance of overcoming synthetic challenges for therapeutic development.


Sujet(s)
Peptides cycliques , Peptides cycliques/composition chimique , Peptides cycliques/synthèse chimique , Cyclisation , Récepteur mu/métabolisme , Oligopeptides/composition chimique , Humains , Acides aminés/composition chimique
12.
Bioorg Chem ; 149: 107507, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38850778

RÉSUMÉ

Opioids are currently the most effective and widely used painkillers in the world. Unfortunately, the clinical use of opioid analgesics is limited by serious adverse effects. Many researchers have been working on designing and optimizing structures in search of novel µ opioid receptor(MOR) agonists with improved analgesic activity and reduced incidence of adverse effects. There are many strategies to develop MOR drugs, mainly focusing on new low efficacy agonists (potentially G protein biased agonists), MOR agonists acting on different Gα subtype, targeting opioid receptors in the periphery, acting on multiple opioid receptor, and targeting allosteric sites of opioid receptors, and others. This review summarizes the design methods, clinical applications, and structure-activity relationships of small-molecule agonists for MOR based on these different design strategies, providing ideas for the development of safer novel opioid ligands with therapeutic potential.


Sujet(s)
Analgésiques morphiniques , Récepteur mu , Récepteur mu/agonistes , Récepteur mu/métabolisme , Humains , Relation structure-activité , Analgésiques morphiniques/pharmacologie , Analgésiques morphiniques/composition chimique , Animaux , Structure moléculaire
13.
Sci Rep ; 14(1): 13507, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38867062

RÉSUMÉ

Traumatic Brain Injury (TBI) induces neuroinflammatory response that can initiate epileptogenesis, which develops into epilepsy. Recently, we identified anti-convulsive effects of naltrexone, a mu-opioid receptor (MOR) antagonist, used to treat drug addiction. While blocking opioid receptors can reduce inflammation, it is unclear if post-TBI seizures can be prevented by blocking MORs. Here, we tested if naltrexone prevents neuroinflammation and/or seizures post-TBI. TBI was induced by a modified Marmarou Weight-Drop (WD) method on 4-week-old C57BL/6J male mice. Mice were placed in two groups: non-telemetry assessing the acute effects or in telemetry monitoring for interictal events and spontaneous seizures both following TBI and naltrexone. Molecular, histological and neuroimaging techniques were used to evaluate neuroinflammation, neurodegeneration and fiber track integrity at 8 days and 3 months post-TBI. Peripheral immune responses were assessed through serum chemokine/cytokine measurements. Our results show an increase in MOR expression, nitro-oxidative stress, mRNA expression of inflammatory cytokines, microgliosis, neurodegeneration, and white matter damage in the neocortex of TBI mice. Video-EEG revealed increased interictal events in TBI mice, with 71% mice developing post-traumatic seizures (PTS). Naltrexone treatment ameliorated neuroinflammation, neurodegeneration, reduced interictal events and prevented seizures in all TBI mice, which makes naltrexone a promising candidate against PTS, TBI-associated neuroinflammation and epileptogenesis in a WD model of TBI.


Sujet(s)
Lésions traumatiques de l'encéphale , Modèles animaux de maladie humaine , Souris de lignée C57BL , Naltrexone , Neuroprotecteurs , Crises épileptiques , Animaux , Naltrexone/pharmacologie , Mâle , Souris , Crises épileptiques/traitement médicamenteux , Crises épileptiques/étiologie , Lésions traumatiques de l'encéphale/complications , Lésions traumatiques de l'encéphale/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Récepteur mu/métabolisme , Électroencéphalographie , Cytokines/métabolisme
14.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38928357

RÉSUMÉ

Cannabidiol (CBD), a phytocannabinoid, appeared to satisfy several criteria for a safe approach to preventing drug-taking behavior, including opioids. However, most successful preclinical and clinical results come from studies in adult males. We examined whether systemic injections of CBD (10 mg/kg, i.p.) during extinction of oxycodone (OXY, 3 mg/kg, i.p.) induced conditioned place preference (CPP) could attenuate the reinstatement of CPP brought about by OXY (1.5 mg/kg, i.p.) priming in adolescent rats of both sexes, and whether this effect is sex dependent. Accordingly, a priming dose of OXY produced reinstatement of the previously extinguished CPP in males and females. In both sexes, this effect was linked to locomotor sensitization that was blunted by CBD pretreatments. However, CBD was able to prevent the reinstatement of OXY-induced CPP only in adolescent males and this outcome was associated with an increased cannabinoid 1 receptor (CB1R) and a decreased mu opioid receptor (MOR) expression in the prefrontal cortex (PFC). The reinstatement of CCP in females was associated with a decreased MOR expression, but no changes were detected in CB1R in the hippocampus (HIP). Moreover, CBD administration during extinction significantly potentialized the reduced MOR expression in the PFC of males and showed a tendency to potentiate the reduced MOR in the HIP of females. Additionally, CBD reversed OXY-induced deficits of recognition memory only in males. These results suggest that CBD could reduce reinstatement to OXY seeking after a period of abstinence in adolescent male but not female rats. However, more investigation is required.


Sujet(s)
Cannabidiol , Oxycodone , Récepteur cannabinoïde de type CB1 , Récepteur mu , Animaux , Cannabidiol/pharmacologie , Mâle , Femelle , Oxycodone/pharmacologie , Rats , Récepteur cannabinoïde de type CB1/métabolisme , Récepteur mu/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Analgésiques morphiniques/pharmacologie , Conditionnement psychologique/effets des médicaments et des substances chimiques
15.
Molecules ; 29(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731416

RÉSUMÉ

The synthesis of stereochemically pure oximes, amines, saturated and unsaturated cyanomethyl compounds, and methylaminomethyl compounds at the C9 position in 3-hydroxy-N-phenethyl-5-phenylmorphans provided µ-opioid receptor (MOR) agonists with varied efficacy and potency. One of the most interesting compounds, (2-((1S,5R,9R)-5-(3-hydroxyphenyl)-2-phenethyl-2-azabicyclo[3.3.1]nonan-9-yl)acetonitrile), was found to be a potent partial MOR agonist (EC50 = 2.5 nM, %Emax = 89.6%), as determined in the forskolin-induced cAMP accumulation assay. Others ranged in potency and efficacy at the MOR, from nanomolar potency with a C9 cyanomethyl compound (EC50 = 0.85 nM) to its totally inactive diastereomer, and three compounds exhibited weak MOR antagonist activity (the primary amine 3, the secondary amine 8, and the cyanomethyl compound 41). Many of the compounds were fully efficacious; their efficacy and potency were affected by both the stereochemistry of the molecule and the specific C9 substituent. Most of the MOR agonists were selective in their receptor interactions, and only a few had δ-opioid receptor (DOR) or κ-opioid receptor (KOR) agonist activity. Only one compound, a C9-methylaminomethyl-substituted phenylmorphan, was moderately potent and fully efficacious as a KOR agonist (KOR EC50 = 18 nM (% Emax = 103%)).


Sujet(s)
Amines , Oximes , Oximes/composition chimique , Oximes/pharmacologie , Stéréoisomérie , Relation structure-activité , Amines/composition chimique , Amines/pharmacologie , Récepteur mu/métabolisme , Récepteur mu/agonistes , Humains , Animaux , Structure moléculaire , Cellules CHO , Morphinanes/composition chimique , Morphinanes/pharmacologie
16.
Nat Commun ; 15(1): 3544, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38740791

RÉSUMÉ

G-protein-coupled receptors (GPCRs) play pivotal roles in various physiological processes. These receptors are activated to different extents by diverse orthosteric ligands and allosteric modulators. However, the mechanisms underlying these variations in signaling activity by allosteric modulators remain largely elusive. Here, we determine the three-dimensional structure of the µ-opioid receptor (MOR), a class A GPCR, in complex with the Gi protein and an allosteric modulator, BMS-986122, using cryogenic electron microscopy. Our results reveal that BMS-986122 binding induces changes in the map densities corresponding to R1673.50 and Y2545.58, key residues in the structural motifs conserved among class A GPCRs. Nuclear magnetic resonance analyses of MOR in the absence of the Gi protein reveal that BMS-986122 binding enhances the formation of the interaction between R1673.50 and Y2545.58, thus stabilizing the fully-activated conformation, where the intracellular half of TM6 is outward-shifted to allow for interaction with the Gi protein. These findings illuminate that allosteric modulators like BMS-986122 can potentiate receptor activation through alterations in the conformational dynamics in the core region of GPCRs. Together, our results demonstrate the regulatory mechanisms of GPCRs, providing insights into the rational development of therapeutics targeting GPCRs.


Sujet(s)
Cryomicroscopie électronique , Récepteur mu , Récepteur mu/métabolisme , Récepteur mu/composition chimique , Récepteur mu/génétique , Régulation allostérique , Humains , Liaison aux protéines , Sous-unités alpha Gi-Go des protéines G/métabolisme , Sous-unités alpha Gi-Go des protéines G/composition chimique , Sous-unités alpha Gi-Go des protéines G/génétique , Cellules HEK293 , Ligands , Modèles moléculaires , Conformation des protéines
17.
Neurochem Int ; 177: 105764, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38729355

RÉSUMÉ

Increasing evidence supported that oxidative stress induced by herniated lumbar disc played important role in the formation of lumbar disc herniation sciatica (LDHS), however, the neural mechanisms underlying LDHS need further clarification. Endomorphin-2 (EM2) is the endogenous ligand for mu-opioid receptor (MOR), and there is increasing evidence implicating the involvement of spinal EM2 in neuropathic pain. In this study, using an nucleus pulposus implantation induced LDHS rat model that displayed obvious mechanical allodynia, it was found that the expression of EM2 in dorsal root ganglion (DRG) and spinal cord was significantly decreased. It was further found that oxidative stress in DRG and spinal cord was significantly increased in LDHS rats, and the reduction of EM2 in DRG and spinal cord was determined by oxidative stress dominated increment of dipeptidylpeptidase IV activity. A systemic treatment with antioxidant could prevent the forming of mechanical allodynia in LDHS rats. In addition, MOR expression in DRG and spinal cord remained unchanged in LDHS rats. Intrathecal injection of MOR antagonist promoted pain behavior in LDHS rats, and the analgesic effect of intrathecal injection of EM2 was stronger than that of endomorphin-1 and morphine. Taken together, our findings suggest that oxidative stress mediated decrement of EM2 in DRG and spinal cord causes the loss of endogenous analgesic effects and enhances the pain sensation of LDHS.


Sujet(s)
Déplacement de disque intervertébral , Oligopeptides , Stress oxydatif , Rat Sprague-Dawley , Sciatalgie , Animaux , Stress oxydatif/physiologie , Stress oxydatif/effets des médicaments et des substances chimiques , Déplacement de disque intervertébral/métabolisme , Rats , Oligopeptides/pharmacologie , Sciatalgie/métabolisme , Sciatalgie/traitement médicamenteux , Mâle , Moelle spinale/métabolisme , Moelle spinale/effets des médicaments et des substances chimiques , Vertèbres lombales , Ganglions sensitifs des nerfs spinaux/métabolisme , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Récepteur mu/métabolisme
18.
J Med Chem ; 67(11): 9355-9373, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38805667

RÉSUMÉ

Development of more efficacious medications with improved safety profiles to manage and treat multiple forms of pain is a critical element of healthcare. To this end, we have designed and synthesized a novel class of tetracyclic pyridopyrroloquinoxalinone derivatives with analgesic properties. The receptor binding profiles and analgesic properties of these tetracyclic compounds were studied. Systematic optimizations of this novel scaffold culminated in the discovery of the clinical candidate, (6bR,10aS)-8-[3-(4-fluorophenoxy)propyl]-6b,7,8,9,10,10a-hexahydro-1H-pyrido[3',4':4,5]pyrrolo[1,2,3-de]quinoxalin-2(3H)-one (compound 5, ITI-333), which exhibited potent binding affinity to serotonin 5-HT2A (Ki = 8.3 nM) and µ-opioid receptors (MOR, Ki = 11 nM) and moderate affinity to adrenergic α1A (Ki = 28 nM) and dopamine D1 (Ki = 50 nM) receptors. ITI-333 acts as a 5-HT2A receptor antagonist, a MOR partial agonist, and an adrenergic α1A receptor antagonist. ITI-333 exhibited dose-dependent analgesic effects in rodent models of acute pain. Currently, this investigational new drug is in phase I clinical development.


Sujet(s)
Analgésiques , Douleur , Animaux , Humains , Analgésiques/pharmacologie , Analgésiques/composition chimique , Analgésiques/synthèse chimique , Analgésiques/usage thérapeutique , Relation structure-activité , Administration par voie orale , Douleur/traitement médicamenteux , Souris , Mâle , Rats , Découverte de médicament , Rat Sprague-Dawley , Biodisponibilité , Récepteur mu/métabolisme , Récepteur mu/agonistes , Pyridines/composition chimique , Pyridines/pharmacologie , Pyridines/synthèse chimique , Pyridines/usage thérapeutique , Pyridines/pharmacocinétique , Pyrroles/composition chimique , Pyrroles/pharmacologie , Pyrroles/synthèse chimique , Pyrroles/pharmacocinétique
19.
Neuropharmacology ; 255: 110019, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38810926

RÉSUMÉ

The endogenous opioid system has been implicated in alcohol consumption and preference in both humans and animals. The mu opioid receptor (MOR) is expressed on multiple cells in the striatum, however little is known about the contributions of specific MOR populations to alcohol drinking behaviors. The current study used mice with a genetic deletion of MOR in cholinergic cells (ChAT-Cre/Oprm1fl/fl) to examine the role of MORs expressed in cholinergic interneurons (CINs) in home cage self-administration paradigms. Male and female ChAT-Cre/Oprm1fl/fl mice were generated and heterozygous Cre+ (knockout) and Cre- (control) mice were tested for alcohol consumption in two drinking paradigms: limited access "Drinking in the Dark" and intermittent access. Quinine was added to the drinking bottles in the DID experiment to test aversion-resistant, "compulsive" drinking. Nicotine and sucrose drinking were also assessed so comparisons could be made with other rewarding substances. Cholinergic MOR deletion did not influence consumption or preference for ethanol (EtOH) in either drinking task. Differences were observed in aversion-resistance in males with Cre + mice tolerating lower concentrations of quinine than Cre-. In contrast to EtOH, preference for nicotine was reduced following cholinergic MOR deletion while sucrose consumption and preference was increased in Cre+ (vs. Cre-) females. Locomotor activity was also greater in females following the deletion. These results suggest that cholinergic MORs participate in preference for rewarding substances. Further, while they are not required for consumption of alcohol alone, cholinergic MORs may influence the tendency to drink despite negative consequences.


Sujet(s)
Consommation d'alcool , Souris knockout , Quinine , Récepteur mu , Récompense , Animaux , Récepteur mu/génétique , Récepteur mu/métabolisme , Mâle , Femelle , Souris , Quinine/pharmacologie , Quinine/administration et posologie , Consommation d'alcool/génétique , Consommation d'alcool/psychologie , Nicotine/pharmacologie , Éthanol/pharmacologie , Éthanol/administration et posologie , Neurones cholinergiques/effets des médicaments et des substances chimiques , Neurones cholinergiques/physiologie , Neurones cholinergiques/métabolisme , Autoadministration , Saccharose/administration et posologie , Apprentissage par évitement/effets des médicaments et des substances chimiques , Apprentissage par évitement/physiologie , Interneurones/effets des médicaments et des substances chimiques , Interneurones/physiologie , Interneurones/métabolisme
20.
Behav Brain Res ; 469: 115065, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38782097

RÉSUMÉ

Acetaminophen (paracetamol) is one of the most popular analgesics for the management of fever and pain but few reports have investigated its antidepressant-like effect. Moreover, the role of the opioidergic pathway has been indicated in depression pathophysiology. This study aimed to examine the involvement of the opioid receptors in the antidepressant-like effect of acetaminophen after acute and sub-chronic administration using mice forced swimming test (FST). Our finding showed that administration of acetaminophen (50 and 100 mg/kg, i.p.) 30 min before the FST produced an antidepressant effect which was reduced by naloxone (1 mg/kg, i.p., a nonselective opioid receptor antagonist). Moreover, we observed that acetaminophen in higher doses (200 and 400 mg/kg) was ineffective. Also, the response of the non-effective dose of acetaminophen (25 mg/kg) was potentiated by the non-effective dose of morphine (0.1 mg/kg) in the FST that was antagonized by naloxone. Also, in contrast to morphine (10 mg/kg), acetaminophen (100 mg/kg, i.p.) induced neither tolerance to the anti-immobility behavior nor withdrawal syndrome after repeated administration. In addition, RT-PCR showed that hippocampal mu- and kappa-opioid receptor mRNA expression increased in mice after repeated administration of acetaminophen; however, morphine therapy for 6 days did not affect kappa-opioid receptor expression. Our findings demonstrated that acetaminophen in lower doses but not high doses revealed an antidepressant-like activity without inducing tolerance and withdrawal syndromes. Moreover, the observed effect of acetaminophen may be via altering the opioid system, particularly hippocampal mu- and kappa-receptors.


Sujet(s)
Acétaminophène , Antidépresseurs , Relation dose-effet des médicaments , Naloxone , Antagonistes narcotiques , Animaux , Acétaminophène/pharmacologie , Acétaminophène/administration et posologie , Mâle , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/administration et posologie , Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Antagonistes narcotiques/administration et posologie , Natation , Dépression/traitement médicamenteux , Dépression/métabolisme , Morphine/pharmacologie , Morphine/administration et posologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Modèles animaux de maladie humaine , Analgésiques morphiniques/pharmacologie , Analgésiques morphiniques/administration et posologie , Analgésiques non narcotiques/pharmacologie , Analgésiques non narcotiques/administration et posologie , Récepteurs aux opioïdes/métabolisme , Récepteurs aux opioïdes/effets des médicaments et des substances chimiques , Récepteur mu/métabolisme , Récepteur mu/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...