Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 600
Filter
1.
Mol Med Rep ; 25(2)2022 Feb.
Article in English | MEDLINE | ID: mdl-34878160

ABSTRACT

The κ­opioid receptor (KOR) is one of the primary receptors of opioids and serves a vital role in the regulation of pain, anesthesia, addiction and other pathological and physiological processes. KOR is associated with several types of cancer and may influence cancer progression. It has been proposed that KOR may represent a new tumor molecular marker and provide a novel basis for molecular targeted therapies for cancer. However, the association between KOR and cancer remains to be explored comprehensively. The present review introduces KOR and its association with different types of cancer. Improved understanding of KOR may facilitate development of novel antitumor therapies.


Subject(s)
Analgesics, Opioid/metabolism , Hypoxia/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic , Receptors, Opioid, kappa/physiology , Animals , Disease Progression , Humans , Receptors, Opioid, kappa/chemistry
2.
Psychopharmacology (Berl) ; 237(12): 3715-3728, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32894343

ABSTRACT

RATIONALE: Major depressive disorder is a leading cause of disability worldwide and is likely precipitated by chronic stress. Although many antidepressants are currently available, these drugs require weeks to months of daily administration before reduction of symptoms occurs and many patients remain treatment-resistant despite several courses of treatment. There is a pressing need for new treatments for stress-related disorders. Kappa opioid receptors (KORs) are a promising new therapeutic target for major depressive disorder and anhedonia because acute KOR blockade prevents many effects of stress in rodents. OBJECTIVES: The following study assessed whether repeated treatment with the selective KOR antagonist aticaprant (also known as JNJ-67953964, and previously LY-2456302 and CERC-501) was effective in reversing behaviors in rodents following exposure to unpredictable chronic mild stress (UCMS). METHODS: Adult male C57BL/6J mice were exposed to 4 weeks of UCMS. After 3 weeks of stress, aticaprant (10 mg/kg) was administered daily for 11 treatments. Behavioral assessments included the sucrose preference test, nesting, forced swim test, hot plate test, light-dark test, and social interaction test. RESULTS: Aticaprant significantly reversed stress-induced deficits produced by UCMS on the SPT, nesting, FST, and hot plate test. The effects of aticaprant persisted through a stress and treatment recovery period. Aticaprant was not effective at reversing behavioral effects caused by stress in the light-dark and social interaction tests. CONCLUSIONS: The results support further study of the role of KORs in regulating circuits related to reward, self-care, and cognition when they are disrupted by chronic stress. They are also consistent with the clinical development of aticaprant as a therapeutic for stress-related disorders targeted at anhedonia, such as depression and post-traumatic stress disorder.


Subject(s)
Benzamides/therapeutic use , Narcotic Antagonists/therapeutic use , Pyrrolidines/therapeutic use , Receptors, Opioid, kappa/antagonists & inhibitors , Stress, Psychological/drug therapy , Stress, Psychological/psychology , Animals , Benzamides/pharmacology , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Narcotic Antagonists/pharmacology , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/physiology , Swimming/psychology
3.
Psychopharmacology (Berl) ; 237(7): 2075-2087, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32372348

ABSTRACT

RATIONALE: Kappa-opioid receptor (KOR) agonists are antinociceptive but have side effects that limit their therapeutic utility. New KOR agonists have been developed that are fully efficacious at the KOR but may produce fewer or reduced side effects that are typical of KOR agonists. OBJECTIVES: We determined behavioral profiles for typical and atypical KOR agonists purported to differ in intracellular-signaling profiles as well as a mu-opioid receptor (MOR) agonist, oxycodone, using a behavioral scoring system based on Novak et al. (Am J Primatol 28:124-138, 1992, Am J Primatol 46:213-227, 1998) and modified to quantify drug-induced effects (e.g., Duke et al. J Pharmacol Exp Ther 366:145-157, 2018). METHODS: Six adult male rhesus monkeys were administered a range of doses of the typical KOR agonists, U50-488H (0.0032-0.1 mg/kg) and salvinorin A (0.00032-0.01 mg/kg); the atypical KOR agonists, nalfurafine (0.0001-0.001 mg/kg) and triazole 1.1 (0.01-0.32 mg/kg); the MOR agonist, oxycodone (0.0032-0.32 mg/kg); and as controls, cocaine (0.032-0.32 mg/kg) and ketamine (0.32-10 mg/kg). For time-course determinations, the largest dose of each KOR agonist or MOR agonist was administered across timepoints (10-320 min). In mixture conditions, oxycodone (0.1 mg/kg) was followed by KOR-agonist administration. RESULTS: Typical KOR agonists produced sedative-like and motor-impairing effects. Nalfurafine was similar to typical KOR agonists on most outcomes, and triazole 1.1 produced no effects on its own except for reducing scratch during time-course determinations. In the mixture, all KOR agonists reduced oxycodone-induced scratching, U50-488H and nalfurafine reduced species-typical activity, and U50-488H increased rest/sleep posture. CONCLUSIONS: Atypical "biased" KOR agonists produce side-effect profiles that are relatively benign (triazole 1.1) or reduced (nalfurafine) compared to typical KOR agonists.


Subject(s)
Analgesics, Opioid/pharmacology , Motor Activity/drug effects , Motor Activity/physiology , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/physiology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Diterpenes, Clerodane/pharmacology , Dose-Response Relationship, Drug , Macaca mulatta , Male , Morphinans/pharmacology , Oxycodone/pharmacology , Spiro Compounds/pharmacology
4.
Psychopharmacology (Berl) ; 237(8): 2405-2418, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32435819

ABSTRACT

RATIONALE: Agonists of the kappa opioid receptor (KOR) have been shown to block the rewarding effects of drugs of abuse, but with negative side effects. The antipruritic drug nalfurafine, approved in Japan in 2009, is a potent, selective KOR agonist that does not cause significant side effects in humans. Nalfurafine has not been extensively tested for its effect on drug reward and reinforcement in preclinical models. OBJECTIVES: The goal of this study was to compare the effects of nalfurafine and a reference KOR agonist for a variety of KOR-mediated endpoints in male C57BL6 mice. Specifically, we aimed to evaluate the "therapeutic window"-doses of agonists lower than those eliciting negative side effects, while still effective for desired therapeutic effects. METHODS: In this study, several low doses of nalfurafine and U50,488 were tested for serum prolactin release, rotarod-mediated sedation, and place-conditioning in male C57BL6 mice. These agonists were also tested for effects on intravenous cocaine self-administration, both on an FR1 schedule and on a progressive ratio schedule for 0.5 mg/kg/infusion cocaine. RESULTS: Serum prolactin levels increased following doses of both nalfurafine (3 µg/kg and 10 µg/kg) and U50,488 (3 mg/kg). These doses did not cause sedation in the rotarod assay or aversion in a place-conditioning assay, but blocked conditioned place preference for cocaine. Immediate pretreatment of mice with 10 µg/kg nalfurafine and 3 mg/kg U50,488, however, potentiated cocaine self-administration. Further 10 µg/kg nalfurafine was also observed to potentiate cocaine-seeking behavior as demonstrated by increased progressive ratio break point. CONCLUSIONS: Both nalfurafine and U50,488 showed a separation of negative side effects and the modulation of cocaine reward, suggesting this effect of KOR agonists at low doses may be characteristic of the KOR system in general. At higher doses, nalfurafine had similar effects to traditional KOR agonists like U50,488, indicating that its relative potency, rather than differences in KOR signaling, may be responsible for its unique effects in humans.


Subject(s)
Behavior, Addictive/drug therapy , Cocaine/administration & dosage , Morphinans/administration & dosage , Receptors, Opioid, kappa/agonists , Reward , Spiro Compounds/administration & dosage , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Behavior, Addictive/psychology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Receptors, Opioid, kappa/physiology , Self Administration , Signal Transduction/drug effects , Signal Transduction/physiology
5.
Bull Exp Biol Med ; 168(6): 727-729, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32337663

ABSTRACT

We studied the role of opioid receptor subtypes in improvement of the functional state of the heart during reperfusion after adaptation to continuous normobaric hypoxia. To this end, male Wistar rats were subjected to continuous normobaric hypoxia (12% O2). Then, the hearts were isolated and exposed to total 45-min ischemia followed by 30-min reperfusion. Opioid receptor antagonists were added to the perfusion solution prior to ischemia. It was found that continuous normobaric hypoxia reduced the release of creatine phosphokinase into the effluent, increased myocardial contractile force, and decreased the end-diastolic pressure during reperfusion; these positive effects were related to activation of cardiac δ2- and µ-opioid receptors.


Subject(s)
Hypoxia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/physiology , Adaptation, Physiological/drug effects , Animals , Benzylidene Compounds/pharmacology , Creatine Kinase/metabolism , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Oligopeptides/pharmacology , Organ Culture Techniques , Peptides , Rats , Rats, Wistar , Receptors, Opioid, mu/antagonists & inhibitors , Tetrahydroisoquinolines/pharmacology
6.
Brain Res ; 1735: 146742, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32114059

ABSTRACT

The dynorphin / kappa opioid receptor (KOR) system has been implicated in many aspects that influence neuropsychiatric disorders. Namely, this system modulates neural circuits that primarily regulate reward seeking, motivation processing, stress responsivity, and pain sensitivity, thus affecting the development of substance and alcohol use disorder (AUD). The effects of this system are often bidirectional and depend on projection targets. To date, a majority of the studies focusing on this system have examined the KOR function using agonists and antagonists. Indeed, there are studies that have examined prodynorphin and dynorphin levels by measuring mRNA and tissue content levels; however, static levels of the neuropeptide and its precursor do not explain complete and online function of the peptide as would be explained by measuring dynorphin transmission in real time. New and exciting methods using optogenetics, chemogenetics, genetic sensors, fast scan cyclic voltammetry are now being developed to detect various neuropeptides with a focus on opioid peptides, including dynorphin. In this review we discuss studies that examine dynorphin projections in areas involved in AUD, its functional involvement in AUD and vulnerability to develop AUD at various ages. Moreover, we discuss dynorphin's role in promoting AUD by dysregulation motivation circuits and how advancements in opioid peptide detection will further our understanding.


Subject(s)
Alcoholism/drug therapy , Dynorphins/pharmacology , Dynorphins/therapeutic use , Alcoholism/metabolism , Animals , Dynorphins/metabolism , Humans , Motivation/drug effects , Neuropeptides/analysis , Neuropeptides/chemistry , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/physiology , Reward
7.
Neuropharmacology ; 165: 107831, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31870854

ABSTRACT

As an integrative hub, the insular cortex (IC) translates external cues into interoceptive states that generate complex physiological, affective, and behavioral responses. However, the precise circuit and signaling mechanisms in the IC that modulate these processes are unknown. Here, we describe a midbrain-projecting microcircuit in the medial aspect of the agranular IC that signals through the Gαi/o-coupled kappa opioid receptor (KOR) and its endogenous ligand dynorphin (Dyn). Within this microcircuit, Dyn is robustly expressed in layer 2/3, while KOR is localized to deep layer 5, which sends a long-range projection to the substantia nigra (SN). Using ex vivo electrophysiology, we evaluated the functional impact of KOR signaling in layer 5 of the IC. We found that bath application of dynorphin decreased GABA release and increased glutamate release on IC-SN neurons, but did not alter their excitability. Conversely, dynorphin decreased the excitability of GABA neurons without altering synaptic transmission. Pretreatment with the KOR antagonist nor-BNI blocked the effects of dynorphin in IC-SN neurons and GABA neurons, indicating that the changes in synaptic transmission and excitability were selectively mediated through KOR. Selective inhibition of IC GABA neurons using a KOR-derived DREADD recapitulated these effects. This work provides insight into IC microcircuitry and indicates that Dyn/KOR signaling may act to directly reduce activity of layer 5 GABA neurons. In turn, KOR-driven inhibition of GABA promotes disinhibition of IC-SN neurons, which can modulate downstream circuits. Our findings present a potential mechanism whereby chronic upregulation of IC Dyn/KOR signaling can lead to altered subcortical function and downstream activity.


Subject(s)
Cerebral Cortex/physiology , GABAergic Neurons/physiology , Receptors, Opioid, kappa/physiology , Substantia Nigra/physiology , Synaptic Transmission , Animals , Female , Male , Mice
8.
Vitam Horm ; 111: 227-246, 2019.
Article in English | MEDLINE | ID: mdl-31421702

ABSTRACT

The organization of estrogenic signaling in the CNS is exceedingly complex. It is comprised of peripherally and centrally synthesized estrogens, and a plethora of types of estrogen receptor that can localize to both the nucleus and the plasma membrane. Moreover, CNS estrogen receptors can exist independent of aromatase (aka estrogen synthase) as well as oligomerize with it, along with a host of other membrane signaling proteins. This ability of CNS estrogen receptors to either to physically pair or exist separately enables locally produced estrogens to act on multiple spatial levels, with a high degree of gradated regulation and plasticity, signaling either in-phase or out-of phase with circulating estrogens. This complexity explains the numerous contradictory findings regarding sex-dependent pain processing and sexually dimorphic opioid antinociception. This review highlights the increasing awareness that estrogens are major endogenous arbiters of both opioid analgesic actions and the mechanisms used to achieve them. This behooves us to understand, and possibly intercede at, the points of intersection of estrogenic signaling and opioid functionality. Factors that integrate estrogenic actions at subcellular, synaptic, and CNS regional levels are likely to be prime drug targets for novel pharmacotherapies designed to modulate CNS estrogen-dependent opioid functionalities and possibly circumvent the current opioid epidemic.


Subject(s)
Analgesics, Opioid/pharmacology , Estrogens/physiology , Reproduction/physiology , Sex Characteristics , Analgesia , Animals , Aromatase , Brain/physiology , Dynorphins/physiology , Female , Humans , Male , Neurosecretory Systems/physiology , Nociception/drug effects , Nociception/physiology , Receptors, Estrogen/physiology , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/physiology , Signal Transduction/physiology
9.
Vitam Horm ; 111: 281-297, 2019.
Article in English | MEDLINE | ID: mdl-31421704

ABSTRACT

Adult oligodendrocyte precursor cells (OPCs) maintain the abilities to differentiate and myelinate denuded axons in demyelinating diseases, such as Multiple Sclerosis (MS), albert often inefficiently. Remyelination therapies seek to enhance endogenous remyelination and represent a promising approach to achieve functional and cellular architectural recovery against neuronal deficits. Recent findings indicate that the kappa opioid receptor (KOR), a G-protein coupled receptor (GPCR), plays an important role in regulating oligodendrocyte differentiation and myelination. In this chapter, we reviewed (1) current knowledge of the functional importance of remyelination in demyelination diseases; (2) the opioids that can alter oligodendroglial proliferation and differentiation; (3) the endogenous KOR signaling in regulating oligodendrocyte myelination.


Subject(s)
Oligodendroglia/physiology , Receptors, Opioid, kappa/physiology , Remyelination/physiology , Animals , Axons/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Demyelinating Diseases/physiopathology , Demyelinating Diseases/therapy , Humans , Myelin Sheath/physiology , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/physiology , Signal Transduction/physiology
10.
Neurochem Int ; 129: 104504, 2019 10.
Article in English | MEDLINE | ID: mdl-31301327

ABSTRACT

Neural circuits that enable an organism to protect itself by promoting escape from immediate threat and avoidance of future injury are conceptualized to carry an "aversive" signal. One of the key molecular elements of these circuits is the kappa opioid receptor (KOR) and its endogenous peptide agonist, dynorphin. In many cases, the aversive response to an experimental manipulation can be eliminated by selective blockade of KOR function, indicating its necessity in transmitting this signal. The dopamine system, through its contributions to reinforcement learning, is also involved in processing of aversive stimuli, and KOR control of dopamine in the context of aversive behavioral states has been intensely studied. In this review, we have discussed the multiple ways in which the KORs regulate dopamine dynamics with a central focus on dopamine neurons and projections from the ventral tegmental area. At the neuronal level, KOR agonists inhibit dopamine neurons both in the somatodendritic region as well as at terminal release sites, through various signaling pathways and ion channels, and these effects are specific to different synaptic sites. While the dominant hypotheses are that aversive states are driven by decreases in dopamine and increases in dynorphin, reported exceptions to these patterns indicate these ideas require refinement. This is critical given that KOR is being considered as a target for development of new therapeutics for anxiety, depression, pain, and other psychiatric disorders.


Subject(s)
Avoidance Learning/physiology , Dopamine/physiology , Dopaminergic Neurons/physiology , Receptors, Opioid, kappa/physiology , Amygdala/metabolism , Animals , Avoidance Learning/drug effects , Dynorphins/physiology , Forecasting , Learning/physiology , Nucleus Accumbens/metabolism , Punishment , Reinforcement, Psychology , Signal Transduction/physiology , Ventral Tegmental Area/metabolism
11.
Behav Pharmacol ; 30(6): 463-470, 2019 09.
Article in English | MEDLINE | ID: mdl-30724802

ABSTRACT

Pain is a significant public health problem, and assessment of pain-related impairment of behavior is a key clinical indicator and treatment target. Similar to opioids and NSAIDs, dopamine (DA) transporter inhibitors block pain-related depression of intracranial self-stimulation (ICSS) in rats. The primary goal of the present study was to determine if the effects of monoamine uptake inhibitors on pain-related depression of ICSS in rats extend to an assay of pain-related depression of nesting in mice. We hypothesized that the DA transporter-selective uptake inhibitor bupropion would block depression of nesting behavior produced by intraperitoneal injection of lactic acid, whereas selective serotonin transporter-selective citalopram, norepinephrine transporter-selective nisoxetine, and the mixed action selective serotonin transporter/norepinephrine transporter inhibitor milnacipran would be ineffective. Effects of the NSAID ketoprofen were also obtained to facilitate interpretation of the effects of the monoamine uptake inhibitors. Consistent with previous findings, ketoprofen blocked pain-related depression of nesting. In contrast, none of the monoamine uptake inhibitors blocked pain-related depression of nesting, although they all blocked pain-related stimulation of stretching. Unlike findings from studies of pain-related depression of ICSS, these results do not support consideration of DA uptake inhibitors for treatment of pain-related depression of behavior.


Subject(s)
Behavior, Animal/drug effects , Nesting Behavior/drug effects , Pain/drug therapy , Analgesics, Opioid/pharmacology , Animals , Bupropion/pharmacology , Citalopram/pharmacology , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Ketoprofen/pharmacology , Lactic Acid/pharmacology , Male , Mice , Mice, Inbred ICR , Milnacipran/pharmacology , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/physiology , Self Stimulation/drug effects
12.
Brain Res ; 1713: 91-101, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30244022

ABSTRACT

Negative affective states are prevalent symptoms in a plethora of neuropsychiatric disorders, including depression and drug addiction. Dysfunction of mesocorticolimbic dopamine systems has been implicated in negative affective states in neuropsychiatric disorders. The dynorphin/kappa-opioid receptor system is a powerful effector of stress-related behavior and is highly enriched within the mesocorticolimbic dopamine system. Dysfunction of dynorphin/KOR signaling within the mesocorticolimbic dopamine system is implicated in promoting symptoms in neuropsychiatric disorders. As such, the kappa-opioid receptor system provides an important therapeutic target to treat negative affective states associated with psychiatric disorders. In this review, we provide a comprehensive overview of the dynorphin/kappa-opioid receptor system and its role in regulating the mesocorticolimbic dopamine system, motivation, and emotional behavior. Furthermore, we highlight unresolved issues in the field and offer some insights for future research.


Subject(s)
Affect/physiology , Dopamine/metabolism , Dynorphins/metabolism , Receptors, Opioid, kappa/metabolism , Affect/drug effects , Animals , Dopamine/physiology , Dynorphins/physiology , Emotions , Humans , Mental Disorders/metabolism , Mental Disorders/physiopathology , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Receptors, Opioid, kappa/physiology , Signal Transduction
13.
Nat Rev Neurosci ; 19(8): 499-514, 2018 08.
Article in English | MEDLINE | ID: mdl-29934561

ABSTRACT

Drug addiction is a worldwide societal problem and public health burden, and results from recreational drug use that develops into a complex brain disorder. The opioid system, one of the first discovered neuropeptide systems in the history of neuroscience, is central to addiction. Recently, opioid receptors have been propelled back on stage by the rising opioid epidemics, revolutions in G protein-coupled receptor research and fascinating developments in basic neuroscience. This Review discusses rapidly advancing research into the role of opioid receptors in addiction, and addresses the key questions of whether we can kill pain without addiction using mu-opioid-receptor-targeting opiates, how mu- and kappa-opioid receptors operate within the neurocircuitry of addiction and whether we can bridge human and animal opioid research in the field of drug abuse.


Subject(s)
Brain/physiopathology , Pain/drug therapy , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/physiology , Substance-Related Disorders/physiopathology , Animals , Humans , Neurons/physiology , Reward , Substance-Related Disorders/etiology
14.
Biol Pharm Bull ; 41(6): 957-960, 2018.
Article in English | MEDLINE | ID: mdl-29863085

ABSTRACT

Olfactory bulbectomized (OBX) mice are characterized by impaired performance in the passive avoidance test and decreased number of cholinergic neurons in the hippocampus. Several studies have reported that κ-opioid receptor agonists improve cognitive function in mice. However, their influence on OBX-induced cognitive dysfunction remains unclear. To address this question, we evaluated the effects of the endogenous κ-opioid receptor agonist dynorphin A (Dyn A) and the selective agonist trans-(-)-U-50488 on the behavior of OBX mice in the passive avoidance test. The cognitive dysfunction of OBX mice was significantly recovered by the intracerebroventricular administration of Dyn A or trans-(-)-U-50488. The effects of these two agonists were counteracted by the selective κ-opioid receptor antagonist nor-binaltorphimine or the inhibitor of acetylcholine release ß-bungarotoxin. These findings suggest that κ-opioid receptor agonists produce anti-dementia effects through activation of cholinergic neurons in OBX mice.


Subject(s)
Cognitive Dysfunction/drug therapy , Memory Disorders/drug therapy , Receptors, Opioid, kappa/agonists , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/therapeutic use , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Cognitive Dysfunction/physiopathology , Dynorphins/pharmacology , Dynorphins/therapeutic use , Hippocampus/physiology , Male , Memory Disorders/physiopathology , Mice , Olfactory Bulb/surgery , Receptors, Opioid, kappa/physiology
15.
Psychopharmacology (Berl) ; 235(5): 1581-1591, 2018 05.
Article in English | MEDLINE | ID: mdl-29492614

ABSTRACT

RATIONALE: Studies suggest that the κ-opioidergic system becomes overactivated as ethanol use disorders develop. Nalmefene, a currently approved treatment for ethanol use disorders, may also elicit some of its main effects via the κ-opioidergic system. However, the exact role of κ-opioid receptors on regulating ethanol intake and contribution to the development of ethanol addiction remains to be elucidated. OBJECTIVES: The aim of the present study was to clarify the role of accumbal κ-opioid receptors in controlling ethanol intake in alcohol-preferring Alko Alcohol (AA) rats. METHODS: Microinfusions of the long-acting and selective κ-opioid receptor antagonist JDTic (1-15 µg/site) were administered bilaterally into the nucleus accumbens shell of AA rats voluntarily consuming 10% ethanol solution in the intermittent, time-restricted two-bottle choice access paradigm. JDTic (10 mg/kg) was also administered subcutaneously. Both the acute and long-term effects of the treatment on ethanol intake were examined. As a reference, nor-BNI (3 µg/site) was administered intra-accumbally. RESULTS: Systemically administered JDTic decreased ethanol intake significantly 2 days and showed a similar trend 4 days after administration. Furthermore, intra-accumbally administered JDTic showed a weak decreasing effect on ethanol intake long-term but had no acute effects. Intra-accumbal administration of nor-BNI tended to decrease ethanol intake. CONCLUSIONS: The results provide further evidence that κ-opioid receptors play a role in controlling ethanol intake and that accumbal κ-opioid receptors participate in the modulation of the reinforcing effects of ethanol. Furthermore, the results suggest that κ-opioid receptor antagonists may be a valuable adjunct in the pharmacotherapy of ethanol use disorders.


Subject(s)
Alcohol Drinking/drug therapy , Ethanol/administration & dosage , Piperidines/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Tetrahydroisoquinolines/administration & dosage , Alcohol Drinking/psychology , Animals , Ethanol/antagonists & inhibitors , Male , Microinjections , Narcotic Antagonists/administration & dosage , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Rats , Receptors, Opioid, kappa/physiology , Reinforcement, Psychology
16.
Mol Pharmacol ; 93(4): 376-386, 2018 04.
Article in English | MEDLINE | ID: mdl-29436492

ABSTRACT

There is abundant evidence for formation of G protein-coupled receptor heteromers in heterologous expression systems, but little is known of the function of heteromers in native systems. Heteromers of δ and κ opioid receptors (DOR-KOR heteromers) have been identified in native systems. We previously reported that activation of DOR-KOR heteromers expressed by rat pain-sensing neurons (nociceptors) produces robust, peripherally mediated antinociception. Moreover, DOR agonist potency and efficacy is regulated by KOR antagonists via allosteric interactions within the DOR-KOR heteromer in a ligand-dependent manner. Here we assessed the reciprocal regulation of KOR agonist function by DOR antagonists in adult rat nociceptors in culture and in a behavioral assay of nociception. Naltrindole enhanced the potency of the KOR agonist 2-(3,4-dichlorophenyl)-N-methyl-N-[(1S)-1-phenyl-2-pyrrolidin-1-ylethyl]acetamide (ICI-199441) 10- to 20-fold, but did not alter responses to 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50488). By contrast, the potency of U50488 was enhanced 20-fold by 7-benzylidenenaltrexone. The efficacy of 6'-guanidinonaltrindole (6'-GNTI) to inhibit nociceptors was blocked by small interfering RNA knockdown of DOR or KOR. Replacing 6'-GNTI occupancy of DOR with either naltrindole or 7-benzylidenenaltrexone abolished 6'-GNTI efficacy. Further, peptides derived from DOR transmembrane segment 1 fused to the cell membrane-penetrating HIV transactivator of transcription peptide also blocked 6'-GNTI-mediated responses ex vivo and in vivo, suggesting that 6'-GNTI efficacy in nociceptors is due to its positive allosteric regulation of KOR via occupancy of DOR in a DOR-KOR heteromer. Together, these results provide evidence for the existence of functional DOR-KOR heteromers in rat peripheral sensory neurons and that reciprocal, ligand-dependent allosteric interactions occur between the DOR and KOR protomers.


Subject(s)
Analgesics, Opioid/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Trigeminal Ganglion/drug effects , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Male , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peripheral Nerves/drug effects , Peripheral Nerves/physiology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/physiology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Trigeminal Ganglion/physiology
17.
Neuropsychopharmacology ; 43(4): 739-750, 2018 03.
Article in English | MEDLINE | ID: mdl-28857070

ABSTRACT

The κ-opioid receptor (KOP-r) system and its endogenous ligands, the dynorphins, are involved in the neurobiological regulation of addictive states, and of mood. There are limited data on the impact of selective KOP-r antagonism in humans on basic biobehavioral functions, or on addictive diseases and mood disorders. Previously studied selective KOP-r antagonists have unusual pharmacodynamic and pharmacokinetic properties (slow development of KOP-r selectivity, extremely long duration of action) that limit translation to human studies. A recently developed selective KOP-r-antagonist, Opra Kappa (LY2456302; CERC-501), has medication-like duration of action, oral bioavailability, and target engagement. The current study is the first investigation of the effects of a KOP-r-antagonist in cocaine-dependent persons in comparison with normal volunteers. In a stress-minimized inpatient setting, we determined the neuroendocrine and neurobehavioral effects of repeated administration of an active dose of Opra Kappa (10 mg p.o. daily, four consecutive days in comparison with an initial baseline day). Healthy volunteers (n=40), persons diagnosed with cocaine dependence in early abstinence (<2 months, EACD) (n=23), and drug-free former cocaine-dependent persons (7-month to 25-year abstinence, DFFCD) (n=7) were studied, with measurements including circulating neuroendocrine hormones, affect, and, in cocaine-dependent persons, cocaine craving. Modest adverse events related to Opra Kappa included pruritus, observed in a subset of individuals. No significant change was observed in serum prolactin levels following Opra Kappa administration, but modest increases in circulating adrenocorticotropic hormone and cortisol were observed. No significant changes were noted in measures of depression or cocaine craving in this stress-minimized setting. Overall, these studies demonstrate that effects of 10 mg Opra Kappa are largely consistent with those predicted for a selective KOP-r antagonist. This medication regimen was tolerable, and is therefore feasible for further studies in cocaine-dependent persons.


Subject(s)
Benzamides/administration & dosage , Cocaine-Related Disorders/diagnosis , Cocaine-Related Disorders/drug therapy , Narcotic Antagonists/administration & dosage , Pyrrolidines/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Administration, Oral , Adult , Behavior, Addictive/diagnosis , Behavior, Addictive/drug therapy , Behavior, Addictive/psychology , Benzamides/adverse effects , Cocaine-Related Disorders/psychology , Female , Humans , Male , Middle Aged , Narcotic Antagonists/adverse effects , Pruritus/chemically induced , Pyrrolidines/adverse effects , Receptors, Opioid, kappa/physiology , Young Adult
18.
Neuropsychopharmacology ; 43(4): 838-850, 2018 03.
Article in English | MEDLINE | ID: mdl-28589966

ABSTRACT

κ-Opioid receptors (KORs) and their endogenous ligand dynorphin are involved in stress-induced alcohol seeking but the mechanisms involved are largely unknown. We previously showed that systemic injections of the KOR agonist U50,488, which induce stress-like aversive states, reinstate alcohol seeking after extinction of the alcohol-reinforced responding. Here, we used the neuronal activity marker Fos and site-specific injections of the KOR antagonist nor-BNI and U50,488 to study brain mechanisms of U50,488-induced reinstatement of alcohol seeking. We trained male Long-Evans rats to self-administer alcohol (12% w/v) for 23-30 days. After extinction of the alcohol-reinforced responding, we tested the effect of U50,488 (0, 1.25, 2.5, and 5 mg/kg) on reinstatement of alcohol seeking. Next, we correlated regional Fos expression with reinstatement induced by the most effective U50,488 dose (5 mg/kg). Based on the correlational Fos results, we determined the effect of bed nucleus of the stria terminalis (BNST) injections of nor-BNI (4 µg/side) on U50,488-induced reinstatement of alcohol seeking, and reinstatement induced by injections of U50,488 (0, 0.3, 1, and 3 µg/side) into the BNST. U50,488-induced reinstatement of alcohol seeking was associated with increased Fos expression in multiple brain areas, including the BNST, where it was significantly correlated with lever pressing. U50,488-induced reinstatement was blocked by BNST nor-BNI injections, and BNST U50,488 injections partially mimicked the drug's systemic effect on reinstatement. Our data indicate that the BNST is a critical site for U50,488-induced reinstatement of alcohol seeking and suggest that KOR/dynorphin mechanisms in this brain area play a key role in stress-induced alcohol seeking.


Subject(s)
Alcohol Drinking/metabolism , Drug-Seeking Behavior/physiology , Ethanol/administration & dosage , Receptors, Opioid, kappa/physiology , Reinforcement, Psychology , Septal Nuclei/metabolism , Alcohol Drinking/psychology , Animals , Drug-Seeking Behavior/drug effects , Male , Narcotic Antagonists/pharmacology , Rats , Rats, Long-Evans , Receptors, Opioid, kappa/antagonists & inhibitors , Self Administration , Septal Nuclei/drug effects
19.
J Oral Facial Pain Headache ; 31(3): 275-284, 2017.
Article in English | MEDLINE | ID: mdl-28738113

ABSTRACT

AIMS: To determine whether estrogen status alters κ-opioid inhibition of nociceptive processing by affecting temporomandibular joint (TMJ) input to neurons in the trigeminal subnucleus caudalis [Vc]/C1-2 region at the spinomedullary junction in female rats. METHODS: TMJ-responsive neurons were recorded in laminae I-II of the Vc/C1-2 region at the spinomedullary junction of ovariectomized female rats treated for 2 days with low-dose estradiol (LE group; 2 mg/day) or high-dose estradiol (HE group; 20 mg/day). Under isoflurane anesthesia, TMJ neurons were activated by adenosine triphosphate (ATP; 1 mM, 20 µl), which was injected into the joint space before and after cumulative doses of a κ-opioid receptor (KOR) agonist (U50488) given systemically (0.03, 0.3, and 3 mg/kg, intravenously) or by local application to the dorsal surface of the Vc/C1-2 region (1 and 10 nmol/30 µl). Analysis of variance and Newman-Keuls test were performed to compare the data. RESULTS: Systemic U50488 caused a dose-related inhibition of ATP-evoked neuronal activity in HE rats and reduced the size of the neuronal cutaneous receptive field (RF), while effects in LE rats were not significant. Systemic U50488 reduced the spontaneous activity of TMJ-responsive neurons to similar levels in LE and HE groups. Locally applied U50488 inhibited ATP-evoked neuronal activity in HE rats, but not in LE rats. Systemic and local administration of the KOR antagonist nor-binaltorphinine (nor-BNI) partially reversed the decrease in Rmag induced by U50488, but had no effect on neurons from LE rats. CONCLUSION: These results indicate that KOR-dependent effects on TMJ-responsive neurons in the superficial laminae of the Vc/C1-2 region in female rats are differentially modified by high and low estrogen status. The site of action for estrogen-induced modulation of TMJ neuronal activity by KOR likely includes second-order neurons in the Vc/C1-2 region.


Subject(s)
Estrogens/physiology , Neurons/physiology , Receptors, Opioid, kappa/physiology , Temporomandibular Joint/physiology , Trigeminal Caudal Nucleus/physiology , Animals , Female , Rats , Rats, Sprague-Dawley
20.
J Neurosci ; 37(32): 7656-7668, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28674176

ABSTRACT

Stress plays a critical role in the neurobiology of mood and anxiety disorders. Sleep and circadian rhythms are affected in many of these conditions. Here we examined the effects of chronic social defeat stress (CSDS), an ethological form of stress, on sleep and circadian rhythms. We exposed male mice implanted with wireless telemetry transmitters to a 10 day CSDS regimen known to produce anhedonia (a depressive-like effect) and social avoidance (an anxiety-like effect). EEG, EMG, body temperature, and locomotor activity data were collected continuously during the CSDS regimen and a 5 day recovery period. CSDS affected numerous endpoints, including paradoxical sleep (PS) and slow-wave sleep (SWS), as well as the circadian rhythmicity of body temperature and locomotor activity. The magnitude of the effects increased with repeated stress, and some changes (PS bouts, SWS time, body temperature, locomotor activity) persisted after the CSDS regimen had ended. CSDS also altered mRNA levels of the circadian rhythm-related gene mPer2 within brain areas that regulate motivation and emotion. Administration of the κ-opioid receptor (KOR) antagonist JDTic (30 mg/kg, i.p.) before CSDS reduced stress effects on both sleep and circadian rhythms, or hastened their recovery, and attenuated changes in mPer2 Our findings show that CSDS produces persistent disruptions in sleep and circadian rhythmicity, mimicking attributes of stress-related conditions as they appear in humans. The ability of KOR antagonists to mitigate these disruptions is consistent with previously reported antistress effects. Studying homologous endpoints across species may facilitate the development of improved treatments for psychiatric illness.SIGNIFICANCE STATEMENT Stress plays a critical role in the neurobiology of mood and anxiety disorders. We show that chronic social defeat stress in mice produces progressive alterations in sleep and circadian rhythms that resemble features of depression as it appears in humans. Whereas some of these alterations recover quickly upon cessation of stress, others persist. Administration of a kappa-opioid receptor (KOR) antagonist reduced stress effects or hastened recovery, consistent with the previously reported antistress effects of this class of agents. Use of endpoints, such as sleep and circadian rhythm, that are homologous across species will facilitate the implementation of translational studies that better predict clinical outcomes in humans, improve the success of clinical trials, and facilitate the development of more effective therapeutics.


Subject(s)
Circadian Rhythm/physiology , Piperidines/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/physiology , Sleep/physiology , Stress, Psychological/physiopathology , Tetrahydroisoquinolines/pharmacology , Animals , Circadian Rhythm/drug effects , Male , Mice , Mice, Inbred C57BL , Piperidines/therapeutic use , Random Allocation , Sleep/drug effects , Social Behavior , Stress, Psychological/drug therapy , Stress, Psychological/psychology , Tetrahydroisoquinolines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL