Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.844
Filter
1.
PeerJ ; 12: e17446, 2024.
Article in English | MEDLINE | ID: mdl-38827306

ABSTRACT

Objectives: To investigate the interaction between tramadol and representative tyrosine kinase inhibitors, and to study the inhibition mode of drug-interaction. Methods: Liver microsomal catalyzing assay was developed. Sprague-Dawley rats were administrated tramadol with or without selected tyrosine kinase inhibitors. Samples were prepared and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was used for analysis. Besides, liver, kidney, and small intestine were collected and morphology was examined by hematoxyline-eosin (H&E) staining. Meanwhile, liver microsomes were prepared and carbon monoxide differential ultraviolet radiation (UV) spectrophotometric quantification was performed. Results: Among the screened inhibitors, crizotinib takes the highest potency in suppressing the metabolism of tramadol in rat/human liver microsome, following non-competitive inhibitory mechanism. In vivo, when crizotinib was co-administered, the AUC value of tramadol increased compared with the control group. Besides, no obvious pathological changes were observed, including cell morphology, size, arrangement, nuclear morphology with the levels of alanine transaminase (ALT) and aspartate transaminase (AST) increased after multiple administration of crizotinib. Meanwhile, the activities of CYP2D1 and CYP3A2 as well as the total cytochrome P450 abundance were found to be decreased in rat liver of combinational group. Conclusions: Crizotinib can inhibit the metabolism of tramadol. Therefore, this recipe should be vigilant to prevent adverse reactions.


Subject(s)
Crizotinib , Cytochrome P-450 CYP3A , Microsomes, Liver , Rats, Sprague-Dawley , Tramadol , Animals , Tramadol/pharmacology , Crizotinib/pharmacology , Rats , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Cytochrome P-450 CYP3A/metabolism , Male , Drug Interactions , Humans , Tandem Mass Spectrometry , Cytochrome P450 Family 2/metabolism , Cytochrome P450 Family 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Analgesics, Opioid/pharmacology
2.
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
4.
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
5.
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
6.
Int J Nanomedicine ; 19: 4759-4777, 2024.
Article in English | MEDLINE | ID: mdl-38828199

ABSTRACT

Background: Opioids are irreplaceable analgesics owing to the lack of alternative analgesics that offer opioid-like pain relief. However, opioids have many undesirable central side effects. Restricting opioids to peripheral opioid receptors could reduce those effects while maintaining analgesia. Methods: To achieve this goal, we developed Tet1-LNP (morphine), a neural-targeting lipid nanoparticle encapsulating morphine that could specifically activate the peripheral opioid receptor in the dorsal root ganglion (DRG) and significantly reduce the side effects caused by the activation of opioid receptors in the brain. Tet1-LNP (morphine) were successfully prepared using the thin-film hydration method. In vitro, Tet1-LNP (morphine) uptake was assessed in differentiated neuron-like PC-12 cells and dorsal root ganglion (DRG) primary cells. The uptake of Tet1-LNP (morphine) in the DRGs and the brain was assessed in vivo. Von Frey filament and Hargreaves tests were used to assess the antinociception of Tet1-LNP (morphine) in the chronic constriction injury (CCI) neuropathic pain model. Morphine concentration in blood and brain were evaluated using ELISA. Results: Tet1-LNP (morphine) had an average size of 131 nm. Tet1-LNP (morphine) showed high cellular uptake and targeted DRG in vitro. CCI mice treated with Tet1-LNP (morphine) experienced prolonged analgesia for nearly 32 h compared with 3 h with free morphine (p < 0.0001). Notably, the brain morphine concentration in the Tet1-LNP (morphine) group was eight-fold lower than that in the morphine group (p < 0.0001). Conclusion: Our study presents a targeted lipid nanoparticle system for peripheral neural delivery of morphine. We anticipate Tet1-LNP (morphine) will offer a safe formulation for chronic neuropathic pain treatment, and promise further development for clinical applications.


Subject(s)
Analgesics, Opioid , Ganglia, Spinal , Morphine , Nanoparticles , Animals , Morphine/administration & dosage , Morphine/pharmacokinetics , Morphine/chemistry , Morphine/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Nanoparticles/chemistry , Rats , PC12 Cells , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Male , Neuralgia/drug therapy , Mice , Lipids/chemistry , Proto-Oncogene Proteins/metabolism , Peripheral Nerves/drug effects , Mixed Function Oxygenases/metabolism , DNA-Binding Proteins , Liposomes
7.
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
8.
Nature ; 630(8015): 141-148, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778097

ABSTRACT

Fentanyl is a powerful painkiller that elicits euphoria and positive reinforcement1. Fentanyl also leads to dependence, defined by the aversive withdrawal syndrome, which fuels negative reinforcement2,3 (that is, individuals retake the drug to avoid withdrawal). Positive and negative reinforcement maintain opioid consumption, which leads to addiction in one-fourth of users, the largest fraction for all addictive drugs4. Among the opioid receptors, µ-opioid receptors have a key role5, yet the induction loci of circuit adaptations that eventually lead to addiction remain unknown. Here we injected mice with fentanyl to acutely inhibit γ-aminobutyric acid-expressing neurons in the ventral tegmental area (VTA), causing disinhibition of dopamine neurons, which eventually increased dopamine in the nucleus accumbens. Knockdown of µ-opioid receptors in VTA abolished dopamine transients and positive reinforcement, but withdrawal remained unchanged. We identified neurons expressing µ-opioid receptors in the central amygdala (CeA) whose activity was enhanced during withdrawal. Knockdown of µ-opioid receptors in CeA eliminated aversive symptoms, suggesting that they mediate negative reinforcement. Thus, optogenetic stimulation caused place aversion, and mice readily learned to press a lever to pause optogenetic stimulation of CeA neurons that express µ-opioid receptors. Our study parses the neuronal populations that trigger positive and negative reinforcement in VTA and CeA, respectively. We lay out the circuit organization to develop interventions for reducing fentanyl addiction and facilitating rehabilitation.


Subject(s)
Dopaminergic Neurons , Fentanyl , Nucleus Accumbens , Receptors, Opioid, mu , Reinforcement, Psychology , Substance Withdrawal Syndrome , Ventral Tegmental Area , Animals , Fentanyl/pharmacology , Receptors, Opioid, mu/metabolism , Mice , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiology , Male , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Substance Withdrawal Syndrome/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Dopamine/metabolism , Optogenetics , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/physiology , Female , Mice, Inbred C57BL , Opioid-Related Disorders/metabolism , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage
9.
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
10.
Neuropharmacology ; 255: 110002, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38754577

ABSTRACT

RATIONALE: Recent studies report that fentanyl analogs with relatively low pKa values produce antinociception in rodents without other mu opioid-typical side effects due to the restriction of their activity to injured tissue with relatively low pH values. However, it is unclear if and to what degree these compounds may produce mu opioid-typical side effects (respiratory depression, reinforcing effects) at doses higher than those required to produce antinociception. OBJECTIVES: The present study compared the inflammatory antinociceptive, respiratory-depressant, and reinforcing effects of fentanyl and two analogs of intermediate (FF3) and low (NFEPP) pKa values in terms of potency and efficacy in male and female Sprague-Dawley rats. METHODS: Nociception was produced by administration of Complete Freund's Adjuvant into the hind paw of subjects, and antinociception was measured using an electronic Von Frey test. Respiratory depression was measured using whole-body plethysmography. Reinforcing effects were measured in self-administration using a progressive-ratio schedule of reinforcement. The dose ranges tested for each drug encompassed no effect to maximal effects. RESULTS: All compounds produced full effects in all measures but varied in potency. FF3 and fentanyl were equipotent in antinociception and self-administration, but FF3 was less potent than fentanyl in respiratory depression. NFEPP was less potent than fentanyl in every measure. The magnitude of potency difference between antinociception and other effects was greater for FF3 than for NFEPP or fentanyl, indicating that FF3 had the widest margin of safety when relating antinociception to respiratory-depressant and reinforcing effects. CONCLUSIONS: Low pKa fentanyl analogs possess potential as safer analgesics, but determining the optimal degree of difference for pKa relative to fentanyl will require further study due to some differences between the current results and findings from prior work with these analogs.


Subject(s)
Analgesics, Opioid , Fentanyl , Rats, Sprague-Dawley , Animals , Fentanyl/pharmacology , Fentanyl/analogs & derivatives , Male , Female , Analgesics, Opioid/pharmacology , Rats , Reinforcement, Psychology , Dose-Response Relationship, Drug , Self Administration , Respiratory Insufficiency/chemically induced , Pain Measurement/drug effects , Pain Measurement/methods
11.
Biomed Pharmacother ; 175: 116665, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701564

ABSTRACT

Opioid receptor agonists are often used when cancer patients undergo surgery or analgesic treatment. As analgesics in clinical care, opioids can provide intraoperative or to chronic cancer pain relief. Immune function plays an important role in anti-cancer therapy, with cellular immunity, comprised principally of T-lymphocytes and natural killer cells, representing the primary anti-cancer immune response. However, it remains unclear whether immune function is further affected with the use of opioids in already immunocompromised cancer patients. This article provides a review of the effects of commonly used clinical opioids, including morphine, oxycodone, fentanyl and tramadol, on immune function in cancer patients. It provides a summary of current evidence regarding the immunomodulatory effects of opioids in the cancer setting and mechanisms underlying these interactions.


Subject(s)
Analgesics, Opioid , Neoplasms , Humans , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/pharmacology , Neoplasms/immunology , Neoplasms/drug therapy , Cancer Pain/drug therapy , Cancer Pain/immunology , Animals , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Immune System/drug effects
12.
Pharmacol Biochem Behav ; 240: 173791, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761993

ABSTRACT

Fentanyl has become the leading driver of opioid overdoses in the United States. Cessation of opioid use represents a challenge as the experience of withdrawal drives subsequent relapse. One of the most prominent withdrawal symptoms that can contribute to opioid craving and vulnerability to relapse is sleep disruption. The endocannabinoid agonist, 2-Arachidonoylglycerol (2-AG), may promote sleep and reduce withdrawal severity; however, the effects of 2-AG on sleep disruption during opioid withdrawal have yet to be assessed. Here, we investigated the effects of 2-AG administration on sleep-wake behavior and diurnal activity in mice during withdrawal from fentanyl. Sleep-wake activity measured via actigraphy was continuously recorded before and after chronic fentanyl administration in both male and female C57BL/6J mice. Immediately following cessation of fentanyl administration, 2-AG was administered intraperitoneally to investigate the impact of endocannabinoid agonism on opioid-induced sleep disruption. We found that female mice maintained higher activity levels in response to chronic fentanyl than male mice. Furthermore, fentanyl administration increased wake and decreased sleep during the light period and inversely increased sleep and decreased wake in the dark period in both sexes. 2-AG treatment increased arousal and decreased sleep in both sexes during first 24-h of withdrawal. On withdrawal day 2, only females showed increased wakefulness with no changes in males, but by withdrawal day 3 male mice displayed decreased rapid-eye movement sleep during the dark period with no changes in female mice. Overall, repeated administration of fentanyl altered sleep and diurnal activity and administration of the endocannabinoid agonist, 2-AG, had sex-specific effects on fentanyl-induced sleep and diurnal changes.


Subject(s)
Arachidonic Acids , Circadian Rhythm , Endocannabinoids , Fentanyl , Glycerides , Mice, Inbred C57BL , Sleep , Substance Withdrawal Syndrome , Animals , Female , Male , Mice , Arachidonic Acids/pharmacology , Glycerides/pharmacology , Fentanyl/pharmacology , Fentanyl/administration & dosage , Circadian Rhythm/drug effects , Sleep/drug effects , Wakefulness/drug effects , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage
13.
Neuropharmacology ; 254: 109972, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38710443

ABSTRACT

Opioid use disorder (OUD) is a chronic condition associated with long-lasting molecular and behavioral changes. Animals with prolonged access to opioids develop behaviors similar to human OUD. Identifying associated molecular changes can provide insight to underpinnings that lead to or maintain OUD. In pilot studies, we identified several miRNA targets that are altered by the administration of oxycodone. We selected mir182 for follow up as it was recently shown to be dysregulated in plasma of men administered oxycodone. In addition, mir182 is increased in reward-related brain regions of male rats following exposure to various addictive substances. The present study utilizes a long-access oxycodone self-administration paradigm to examine changes in mir182 and its mRNA targets associated with neuroplasticity, which may be involved in the maintenance of OUD-like phenotype in rats. Male rats were trained to self-administer oxycodone (0.1 mg/kg/infusion, i. v.) for 6 h daily sessions for 12 days. Each animal had a yoked saline control that received matched saline infusions. Animals were then tested on a progressive ratio schedule to measure motivation to obtain a single infusion of oxycodone. Drug seeking was measured following 28 days of forced abstinence using a 90-min cued/test. RTqPCR was utilized to measure mir182 and mRNA targets related to neuroplasticity (wnt3, plppr4, pou3f3, tle4, cacna2d, and bdnf) from the nucleus accumbens. Data revealed that animals responded on a continuum for oxycodone. When divided into two groups termed high- and low responders, animals diverged during self-administration acquisition and maintained differences in behavior and gene expression throughout the study. mir182 was upregulated in the nucleus accumbens of both high and low responders and negatively correlated with tle4, which showed a strong negative correlation with reinstatement behavior. mRNA target levels were correlated with behaviors associated with increased severity of OUD behavior in male rats.


Subject(s)
MicroRNAs , Neuronal Plasticity , Oxycodone , Self Administration , Animals , Male , Oxycodone/administration & dosage , Oxycodone/pharmacology , Neuronal Plasticity/drug effects , Rats , MicroRNAs/metabolism , MicroRNAs/genetics , Individuality , Rats, Sprague-Dawley , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Opioid-Related Disorders/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics
14.
Eur J Pharmacol ; 975: 176648, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38759706

ABSTRACT

Opioids are used for pain relief in patients suffering from acute myocardial ischemia or infarction. Clinical and laboratory studies demonstrate that morphine treated patients or the experimental animal model suffering acute myocardial ischemia and reperfusion, may worsen myocardial viability. As transient receptor potential vanilloid 1 (TRPV1) plays important roles in pain sensation and cardio-protection, we query whether opioids may exacerbate myocardial viability via interaction with TRPV1 activity in the pain relief. We found the co-expressions of TRPV1 and opioid µ, δ and κ receptors in adult rat cardiomyocytes. Intravenous injection of morphine (0.3 mg/kg) at 20 min after induction of myocardial ischemia, in the rat model of acute myocardial ischemia and reperfusion, induced significant reduction of phosphorylated TRPV1 (p-TRPV1) in the ventricular myocardium and increase in serum cardiac troponin I (cTnI), compared with the ischemia/reperfusion controls (all P < 0.05). The effects of morphine were completely reversed by selective opioid µ, δ and κ receptor antagonists. While significant upregulation of p-TRPV1 (P < 0.05) and improvement of ±dP/dt max (all P < 0.05) were detected in the animals giving the same dose of morphine before induction of myocardial ischemia. The changes in p-TRPV1 correlate with the alterations of cTnI (r = -0.5840, P = 0.0283) and ±dP/dt max (r = 0.8084, P = 0.0005 and r = -0.8133, P = 0.0004, respectively). The findings of this study may indicate that potentiation and attenuation of TRPV1 sensitivity correlate with the improvement of the cardiac performance and the aggravation of myocardial viability, respectively, by giving morphine before and during myocardial ischemia and reperfusion.


Subject(s)
Morphine , Myocardial Reperfusion Injury , Rats, Sprague-Dawley , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Morphine/pharmacology , Phosphorylation/drug effects , Male , Rats , Time Factors , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Analgesics, Opioid/pharmacology , Receptors, Opioid/metabolism , Troponin I/metabolism , Troponin I/blood , Myocardium/metabolism , Myocardium/pathology
15.
Neurosci Biobehav Rev ; 162: 105713, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733895

ABSTRACT

The kappa opioid receptor (KOR) system is implicated in dysphoria and as an "anti-reward system" during withdrawal from opioids. However, no clear consensus has been made in the field, as mixed findings have been reported regarding the relationship between the KOR system and opioid use. This review summarizes the studies to date on the KOR system and opioids. A systematic scoping review was reported following PRISMA guidelines and conducted based on the published protocol. Comprehensive searches of several databases were done in the following databases: MEDLINE, Embase, PsycINFO, Web of Science, Scopus, and Cochrane. We included preclinical and clinical studies that tested the administration of KOR agonists/antagonists or dynorphin and/or measured dynorphin levels or KOR expression during opioid intoxication or withdrawal from opioids. One hundred studies were included in the final analysis. Preclinical administration of KOR agonists decreased drug-seeking/taking behaviors and opioid withdrawal symptoms. KOR antagonists showed mixed findings, depending on the agent and/or type of withdrawal symptom. Administration of dynorphins attenuated opioid withdrawal symptoms both in preclinical and clinical studies. In the limited number of available studies, dynorphin levels were found to increase in cerebrospinal fluid (CSF) and peripheral blood lymphocytes (PBL) of opioid use disorder subjects (OUD). In animals, dynorphin levels and/or KOR expression showed mixed findings during opioid use. The KOR/dynorphin system appears to have a multifaceted and complex nature rather than simply functioning as an anti-reward system. Future research in well-controlled study settings is necessary to better understand the clinical role of the KOR system in opioid use.


Subject(s)
Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/agonists , Humans , Animals , Opioid-Related Disorders/metabolism , Analgesics, Opioid/pharmacology , Dynorphins/metabolism , Substance Withdrawal Syndrome/metabolism
16.
Addict Biol ; 29(5): e13403, 2024 May.
Article in English | MEDLINE | ID: mdl-38735880

ABSTRACT

Synthetic opioids such as fentanyl contribute to the vast majority of opioid-related overdose deaths, but fentanyl use remains broadly understudied. Like other substances with misuse potential, opioids cause lasting molecular adaptations to brain reward circuits, including neurons in the ventral tegmental area (VTA). The VTA contains numerous cell types that play diverse roles in opioid use and relapse; however, it is unknown how fentanyl experience alters the transcriptional landscape in specific subtypes. Here, we performed single nuclei RNA sequencing to study transcriptional programs in fentanyl-experienced mice. Male and female C57/BL6 mice self-administered intravenous fentanyl (1.5 µg/kg/infusion) or saline for 10 days. After 24 h abstinence, VTA nuclei were isolated and prepared for sequencing on the 10× platform. We identified different patterns of gene expression across cell types. In dopamine neurons, we found enrichment of genes involved in growth hormone signalling. In dopamine-glutamate-GABA combinatorial neurons, and some GABA neurons, we found enrichment of genes involved in Pi3k-Akt signalling. In glutamate neurons, we found enrichment of genes involved in cholinergic signalling. We identified transcriptional regulators for the differentially expressed genes in each neuron cluster, including downregulated transcriptional repressor Bcl6, and upregulated transcription factor Tcf4. We also compared the fentanyl-induced gene expression changes identified in mouse VTA with a published rat dataset in bulk VTA, and found overlap in genes related to GABAergic signalling and extracellular matrix interaction. Together, we provide a comprehensive picture of how fentanyl self-administration alters the transcriptional landscape of the mouse VTA that serves as the foundation for future mechanistic studies.


Subject(s)
Analgesics, Opioid , Fentanyl , Mice, Inbred C57BL , Ventral Tegmental Area , Animals , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Mice , Fentanyl/pharmacology , Male , Female , Analgesics, Opioid/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Self Administration , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Neurons/drug effects , Neurons/metabolism , Opioid-Related Disorders/genetics
17.
Drug Alcohol Depend ; 260: 111320, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38723374

ABSTRACT

BACKGROUND: A single administration of the opioid receptor antagonist methocinnamox (MCAM) antagonizes the antinociceptive effects of µ-opioid receptor agonists for 2 weeks or longer. Such a long duration of antagonism could necessitate the use of nonopioid drugs for treating pain in patients receiving MCAM for opioid use disorder (OUD). METHODS: The antinociceptive effects of fentanyl and nonopioid drugs were assessed in 24 male Sprague Dawley rats using a complete Freund's adjuvant (CFA) model of inflammatory pain. Twelve rats received 10mg/kg MCAM and 12 received vehicle; half (n=6) of the animals from each treatment group were treated (intraplantar) with CFA or saline. Hypersensitivity to mechanical stimulation was measured using a von Frey anesthesiometer. Fentanyl (0.01-0.1mg/kg), ketamine (17.8-56mg/kg), gabapentin (32-100mg/kg), meloxicam (3.2-10mg/kg), and ∆9-tetrahydrocannabinol (THC, 1-10mg/kg) were administered intraperitoneally and tested every 3 days in a pseudorandom order. Next, the same drugs were studied for effects on motor performance using a rotarod apparatus. RESULTS: CFA-induced hypersensitivity was attenuated by fentanyl in vehicle- but not MCAM-treated rats. THC, ketamine, and gabapentin attenuated (up to 82, 66, and 46 %, respectively) CFA-evoked mechanical hypersensitivity in both MCAM- and vehicle-treated rats. Meloxicam failed to alter CFA-evoked mechanical hypersensitivity in either group. Fentanyl, THC, gabapentin, and meloxicam did not affect motor performance in either group whereas ketamine impaired motor performance in both groups (up to 71 % reduction in latency to fall). CONCLUSIONS: These data suggest that ketamine, gabapentin, and THC could be effective for treating inflammatory pain under conditions of long term µ-opioid receptor antagonism.


Subject(s)
Analgesics , Fentanyl , Rats, Sprague-Dawley , Animals , Male , Fentanyl/pharmacology , Rats , Analgesics/pharmacology , Gabapentin/pharmacology , Gabapentin/therapeutic use , Narcotic Antagonists/pharmacology , Pain/drug therapy , Analgesics, Opioid/pharmacology , Ketamine/pharmacology , Analgesics, Non-Narcotic/pharmacology , Analgesics, Non-Narcotic/therapeutic use , Freund's Adjuvant , Pain Measurement/drug effects , Pain Measurement/methods , Amines/pharmacology , Amines/therapeutic use , gamma-Aminobutyric Acid , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use
18.
J Neurosci Methods ; 408: 110171, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38777156

ABSTRACT

BACKGROUND: Although the effects on neural activation and glucose consumption caused by opiates such as morphine are known, the metabolic machinery underlying opioid use and misuse is not fully explored. Multiphoton microscopy (MPM) techniques have been developed for optical imaging at high spatial resolution. Despite the increased use of MPM for neural imaging, the use of intrinsic optical contrast has seen minimal use in neuroscience. NEW METHOD: We present a label-free, multimodal microscopy technique for metabolic profiling of murine brain tissue following incubation with morphine sulfate (MSO4). We evaluate two- and three-photon excited autofluorescence, and second and third harmonic generation to determine meaningful intrinsic contrast mechanisms in brain tissue using simultaneous label-free, autofluorescence multi-harmonic (SLAM) microscopy. RESULTS: Regional differences quantified in the cortex, caudate, and thalamus of the brain demonstrate region-specific changes to metabolic profiles measured from FAD intensity, along with brain-wide quantification. While the overall intensity of FAD signal significantly decreased after morphine incubation, this metabolic molecule accumulated near the nucleus accumbens. COMPARISON WITH EXISTING METHODS: Histopathology requires tissue fixation and staining to determine cell type and morphology, lacking information about cellular metabolism. Tools such as fMRI or PET imaging have been widely used, but lack cellular resolution. SLAM microscopy obviates the need for tissue preparation, permitting immediate use and imaging of tissue with subcellular resolution in its native environment. CONCLUSIONS: This study demonstrates the utility of SLAM microscopy for label-free investigations of neural metabolism, especially the intensity changes in FAD autofluorescence and structural morphology from third-harmonic generation.


Subject(s)
Brain , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Morphine , Animals , Morphine/pharmacology , Microscopy, Fluorescence, Multiphoton/methods , Brain/drug effects , Brain/metabolism , Brain/diagnostic imaging , Mice , Male , Analgesics, Opioid/pharmacology , Narcotics/pharmacology
19.
J Med Chem ; 67(11): 9173-9193, 2024 Jun 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
20.
ACS Chem Neurosci ; 15(11): 2091-2098, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38747710

ABSTRACT

Xylazine (also known as "tranq") is a potent nonopioid veterinary sedative that has recently experienced a surge in use as a drug adulterant, most often combined with illicitly manufactured fentanyl. This combination may heighten the risk of fatal overdose. Xylazine has no known antidote approved for use in humans, and age-adjusted overdose deaths involving xylazine were 35 times higher in 2021 than 2018. In April 2023, the Biden Administration declared xylazine-laced fentanyl an emerging drug threat in the United States. In 2022, the Drug Enforcement Agency (DEA) reported nearly a quarter of seized fentanyl powder contained xylazine. This dramatic increase in prevalence has solidified the status of xylazine as an emerging drug of abuse and an evolving threat to public health. The following narrative review outlines the synthesis, pharmacokinetics, pharmacodynamics, and adverse effects of xylazine, as well as the role it may play in the ongoing opioid epidemic.


Subject(s)
Xylazine , Xylazine/pharmacology , Humans , Animals , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/chemistry , Fentanyl/pharmacology , Fentanyl/chemistry , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Drug Overdose/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...