Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.805
Filter
1.
Sci Rep ; 14(1): 16250, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009645

ABSTRACT

[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.


Subject(s)
Brain , Carbon Radioisotopes , Fentanyl , Positron-Emission Tomography , Receptors, Opioid, mu , Animals , Receptors, Opioid, mu/metabolism , Fentanyl/analogs & derivatives , Fentanyl/metabolism , Fentanyl/pharmacology , Rats , Positron-Emission Tomography/methods , Brain/metabolism , Brain/diagnostic imaging , Male , Rats, Sprague-Dawley , Radiopharmaceuticals/pharmacokinetics
2.
Molecules ; 29(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38998913

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Molecular Docking Simulation , Receptors, Opioid, mu , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Animals , Mice , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Analgesics, Opioid/chemical synthesis , Humans , Structure-Activity Relationship , Pain/drug therapy , Male , Molecular Structure , Thiophenes , Urea/analogs & derivatives
3.
Nature ; 631(8021): 686-693, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961287

ABSTRACT

The µ-opioid receptor (µOR) is a well-established target for analgesia1, yet conventional opioid receptor agonists cause serious adverse effects, notably addiction and respiratory depression. These factors have contributed to the current opioid overdose epidemic driven by fentanyl2, a highly potent synthetic opioid. µOR negative allosteric modulators (NAMs) may serve as useful tools in preventing opioid overdose deaths, but promising chemical scaffolds remain elusive. Here we screened a large DNA-encoded chemical library against inactive µOR, counter-screening with active, G-protein and agonist-bound receptor to 'steer' hits towards conformationally selective modulators. We discovered a NAM compound with high and selective enrichment to inactive µOR that enhances the affinity of the key opioid overdose reversal molecule, naloxone. The NAM works cooperatively with naloxone to potently block opioid agonist signalling. Using cryogenic electron microscopy, we demonstrate that the NAM accomplishes this effect by binding a site on the extracellular vestibule in direct contact with naloxone while stabilizing a distinct inactive conformation of the extracellular portions of the second and seventh transmembrane helices. The NAM alters orthosteric ligand kinetics in therapeutically desirable ways and works cooperatively with low doses of naloxone to effectively inhibit various morphine-induced and fentanyl-induced behavioural effects in vivo while minimizing withdrawal behaviours. Our results provide detailed structural insights into the mechanism of negative allosteric modulation of the µOR and demonstrate how this can be exploited in vivo.


Subject(s)
Cryoelectron Microscopy , Morphine , Naloxone , Receptors, Opioid, mu , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Naloxone/pharmacology , Animals , Mice , Allosteric Regulation/drug effects , Humans , Morphine/pharmacology , Morphine/chemistry , Male , Models, Molecular , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Analgesics, Opioid/metabolism , Narcotic Antagonists/pharmacology , Narcotic Antagonists/chemistry , Ligands , Female , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Opiate Overdose/drug therapy , Kinetics , Fentanyl/chemistry , Fentanyl/pharmacology , Fentanyl/analogs & derivatives
4.
Science ; 384(6700): eadn0886, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843332

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Avoidance Learning , Opioid-Related Disorders , Oxycodone , Parabrachial Nucleus , Prefrontal Cortex , Receptors, Opioid, mu , Reward , Animals , Male , Mice , Analgesics, Opioid/pharmacology , Connectome , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Opioid-Related Disorders/metabolism , Oxycodone/pharmacology , Parabrachial Nucleus/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pyramidal Cells/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Substance Withdrawal Syndrome/metabolism , Transcriptome
5.
Sci Rep ; 14(1): 13507, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38867062

ABSTRACT

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.


Subject(s)
Brain Injuries, Traumatic , Disease Models, Animal , Mice, Inbred C57BL , Naltrexone , Neuroprotective Agents , Seizures , Animals , Naltrexone/pharmacology , Male , Mice , Seizures/drug therapy , Seizures/etiology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, Opioid, mu/metabolism , Electroencephalography , Cytokines/metabolism
6.
Mol Pain ; 20: 17448069241260348, 2024.
Article in English | MEDLINE | ID: mdl-38828868

ABSTRACT

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.


Subject(s)
Dinoprostone , Morphine , Nociceptors , Morphine/pharmacology , Animals , Nociceptors/drug effects , Nociceptors/metabolism , Dinoprostone/metabolism , Dinoprostone/pharmacology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Male , Rats , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Rats, Sprague-Dawley , Dose-Response Relationship, Drug
7.
Bioorg Chem ; 149: 107507, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850778

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Humans , Structure-Activity Relationship , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Animals , Molecular Structure
8.
Nat Commun ; 15(1): 5372, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918367

ABSTRACT

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.


Subject(s)
Peptides, Cyclic , Peptides, Cyclic/chemistry , Peptides, Cyclic/chemical synthesis , Cyclization , Receptors, Opioid, mu/metabolism , Oligopeptides/chemistry , Humans , Amino Acids/chemistry
9.
Exp Dermatol ; 33(6): e15096, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38922774

ABSTRACT

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.


Subject(s)
Asian People , Melanins , Receptors, Opioid, delta , Skin Aging , Humans , Female , Melanins/metabolism , Melanins/biosynthesis , Adult , Receptors, Opioid, delta/metabolism , Middle Aged , Young Adult , Epidermis/metabolism , Receptors, Opioid, mu/metabolism , China
11.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928357

ABSTRACT

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.


Subject(s)
Cannabidiol , Oxycodone , Receptor, Cannabinoid, CB1 , Receptors, Opioid, mu , Animals , Cannabidiol/pharmacology , Male , Female , Oxycodone/pharmacology , Rats , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, mu/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Analgesics, Opioid/pharmacology , Conditioning, Psychological/drug effects
12.
J Med Chem ; 67(12): 10447-10463, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38869493

ABSTRACT

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.


Subject(s)
Fentanyl , Receptors, Opioid, mu , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Fentanyl/pharmacology , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Stereoisomerism , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Animals , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Molecular Conformation , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Analgesics, Opioid/chemical synthesis , CHO Cells , Cricetulus
13.
Genes Brain Behav ; 23(3): e12906, 2024 06.
Article in English | MEDLINE | ID: mdl-38861664

ABSTRACT

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.


Subject(s)
Arvicolinae , Maternal Behavior , Nucleus Accumbens , Pair Bond , Receptors, Opioid, mu , Animals , Female , Arvicolinae/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Maternal Behavior/physiology , Nucleus Accumbens/metabolism , Pregnancy , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Gyrus Cinguli/metabolism , Preoptic Area/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism
14.
Adv Neurobiol ; 35: 221-239, 2024.
Article in English | MEDLINE | ID: mdl-38874725

ABSTRACT

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.


Subject(s)
Exercise Therapy , Exercise , Receptors, Opioid, mu , Humans , Receptors, Opioid, mu/metabolism , Exercise/physiology , Exercise Therapy/methods , Depression/therapy , Depression/metabolism , Treatment Outcome
15.
Adv Neurobiol ; 35: 287-313, 2024.
Article in English | MEDLINE | ID: mdl-38874729

ABSTRACT

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.


Subject(s)
Mitragyna , Plant Extracts , Receptors, Opioid, kappa , Humans , Plant Extracts/pharmacology , Animals , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/agonists , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/agonists , Asia, Southeastern
16.
Drugs ; 84(6): 717-720, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38771484

ABSTRACT

Tegileridine () is a small molecule µ-opioid receptor biased agonist developed by Jiangsu Hengrui Pharmaceuticals Co., Ltd for the treatment of postoperative pain. Tegileridine selectively activates the G-protein-coupled pathway, which mediates strong central analgesic effects and only weakly activates the ß-arrestin-2 pathway implicated in adverse events like respiratory depression and gastrointestinal dysfunction. In January 2024, tegileridine received its first approval in China for the treatment of moderate to severe pain after abdominal surgery. This article summarizes the milestones in the development of tegileridine leading to this first approval for the treatment of moderate to severe pain after abdominal surgery.


Subject(s)
Drug Approval , Pain, Postoperative , Receptors, Opioid, mu , Humans , Pain, Postoperative/drug therapy , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Analgesics, Opioid/therapeutic use , China , Thiophenes/pharmacology , Thiophenes/therapeutic use , Thiophenes/adverse effects , Spiro Compounds
17.
Bioorg Chem ; 148: 107489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797065

ABSTRACT

The number of opioid-related overdose deaths and individuals that have suffered from opioid use disorders have significantly increased over the last 30 years. FDA approved maintenance therapies to treat opioid use disorder may successfully curb drug craving and prevent relapse but harbor adverse effects that reduce patient compliance. This has created a need for new chemical entities with improved patient experience. Previously our group reported a novel lead compound, NAT, a mu-opioid receptor antagonist that potently antagonized the antinociception of morphine and showed significant blood-brain barrier permeability. However, NAT belongs to thiophene containing compounds which are known structural alerts for potential oxidative metabolism. To overcome this, 15 NAT derivatives with various substituents at the 5'-position of the thiophene ring were designed and their structure-activity relationships were studied. These derivatives were characterized for their binding affinity, selectivity, and functional activity at the mu opioid receptor and assessed for their ability to antagonize the antinociceptive effects of morphine in vivo. Compound 12 showed retention of the basic pharmacological attributes of NAT while improving the withdrawal effects that were experienced in opioid-dependent mice. Further studies will be conducted to fully characterize compound 12 to examine whether it would serve as a new lead for opioid use disorder treatment and management.


Subject(s)
Receptors, Opioid, mu , Animals , Structure-Activity Relationship , Mice , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/antagonists & inhibitors , Humans , Molecular Structure , Thiophenes/chemistry , Thiophenes/pharmacology , Thiophenes/chemical synthesis , Thiophenes/therapeutic use , Male , Dose-Response Relationship, Drug , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemistry , Narcotic Antagonists/pharmacology , Narcotic Antagonists/chemistry , Morphine/pharmacology
18.
J Med Chem ; 67(11): 9173-9193, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38810170

ABSTRACT

While in the process of designing more effective synthetic opioid rescue agents, we serendipitously identified a new chemotype of potent synthetic opioid. Here, we report that conformational constraint of a piperazine ring converts a mu opioid receptor (MOR) antagonist into a potent MOR agonist. The prototype of the series, which we have termed atoxifent (2), possesses potent in vitro agonist activity. In mice, atoxifent displayed long-lasting antinociception that was reversible with naltrexone. Repeated dosing of atoxifent produced antinociceptive tolerance and a level of withdrawal like that of fentanyl. In rats, while atoxifent produced complete loss of locomotor activity like fentanyl, it failed to produce deep respiratory depression associated with fentanyl-induced lethality. Assessment of brain biodistribution demonstrated ample distribution of atoxifent into the brain with a Tmax of approximately 0.25 h. These results indicate enhanced safety for atoxifent-like molecules compared to fentanyl.


Subject(s)
Analgesics, Opioid , Fentanyl , Receptors, Opioid, mu , Respiratory Insufficiency , Animals , Mice , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/chemistry , Rats , Male , Fentanyl/pharmacology , Fentanyl/chemical synthesis , Fentanyl/chemistry , Structure-Activity Relationship , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/chemical synthesis , Piperazines/therapeutic use , Piperazines/pharmacokinetics , Humans , Rats, Sprague-Dawley , Tissue Distribution , Brain/metabolism , Brain/drug effects , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/chemical synthesis , Naltrexone/chemistry , Naltrexone/therapeutic use
19.
Behav Brain Res ; 469: 115065, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38782097

ABSTRACT

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.


Subject(s)
Acetaminophen , Antidepressive Agents , Dose-Response Relationship, Drug , Naloxone , Narcotic Antagonists , Animals , Acetaminophen/pharmacology , Acetaminophen/administration & dosage , Male , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Swimming , Depression/drug therapy , Depression/metabolism , Morphine/pharmacology , Morphine/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Analgesics, Non-Narcotic/pharmacology , Analgesics, Non-Narcotic/administration & dosage , Receptors, Opioid/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/drug effects
20.
Neurochem Int ; 177: 105764, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38729355

ABSTRACT

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.


Subject(s)
Intervertebral Disc Displacement , Oligopeptides , Oxidative Stress , Rats, Sprague-Dawley , Sciatica , Animals , Oxidative Stress/physiology , Oxidative Stress/drug effects , Intervertebral Disc Displacement/metabolism , Rats , Oligopeptides/pharmacology , Sciatica/metabolism , Sciatica/drug therapy , Male , Spinal Cord/metabolism , Spinal Cord/drug effects , Lumbar Vertebrae , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Receptors, Opioid, mu/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...