Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neurosci ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744530

ABSTRACT

Sleep disorders affect millions of people around the world and have a high comorbidity with psychiatric disorders. While current hypnotics mostly increase non-rapid eye movement sleep (NREMS), drugs acting selectively on enhancing rapid eye movement sleep (REMS) are lacking. This polysomnographic study in male rats showed that the first-in-class selective melatonin MT1 receptor partial agonist UCM871 increases the duration of REMs without affecting that of NREMS. The REMS-promoting effects of UCM871 occurred by inhibiting, in a dose-response manner, the firing activity of the locus coeruleus (LC) norepinephrine (NE) neurons, which express MT1 receptors. The increase of REMS duration and the inhibition of LC-NE neuronal activity by UCM871 were abolished by MT1 pharmacological antagonism and by an adeno-associated viral (AAV) vector which selectively knocked down MT1 receptors in the LC-NE neurons. In conclusion, MT1 receptor agonism inhibits LC-NE neurons and triggers REMS, thus representing a novel mechanism and target for REMS disorders and/or psychiatric disorders associated with REMS impairments.Significance Statement Rapid eye movement sleep (REMS) is involved in the processes of memory consolidation and emotional regulation, but drugs selectively enhancing REMS are scant. Herein, we show that the first-in-class selective melatonin MT1 receptor agonist UCM871, by inhibiting the activity of norepinephrine neurons in the locus coeruleus, an important nucleus regulating the sleep/wake cycle, selectively increases the duration of REMS. These findings enhance our current understanding of the neurobiology and pharmacology of REMS and provide a possible novel mechanism and target for disorders associated with REMS dysfunctions.

2.
Article in English | MEDLINE | ID: mdl-37257771

ABSTRACT

Neuropathic pain (NP) is often treated with opioids, the prolonged use of which causes tolerance to their analgesic effect and can potentially cause death by overdose. The phytocannabinoid delta-9-tetrahydrocannabinol (THC) may be an effective alternative analgesic to treat NP in morphine-tolerant subjects. Male Wistar rats developed NP after spared nerve injury, and were then treated with increasing doses of THC (1, 1.5, 2, 2.5, and 5 mg/kg, intraperitoneally), which reduced mechanical allodynia at the dose of 2.5 and 5 mg/kg. Another group of NP rats were treated with morphine (5 mg/kg, twice daily for 7 days, subcutaneously), until tolerance developed, and on day 8 received a single dose of THC (2.5 mg/kg), which significantly reduced mechanical allodynia. To evaluate the modulation of THC in the descending pain pathway, in vivo electrophysiological recordings of pronociceptive ON cells and antinociceptive OFF cells in the rostroventral medulla (RVM) were recorded after intra-PAG microinjection of THC (10 µg/µl). NP rats with morphine tolerance, compared to the control one, showed a tonic reduction of the spontaneous firing rate of ON cells by 44%, but the THC was able to further decrease it (a hallmark of many analgesic drugs acting at supraspinal level). On the other hand, the firing rate, of the antinociceptive OFF cells was increased after morphine tolerance by 133%, but the THC failed to further activate it. Altogether, these findings indicate that a single dose of THC produces antiallodynic effect in individuals with NP who are tolerant to morphine, acting mostly on the ON cells of the descending pain pathways, but not on OFF cells.


Subject(s)
Morphine , Neuralgia , Rats , Male , Animals , Morphine/pharmacology , Hyperalgesia/drug therapy , Dronabinol/pharmacology , Rats, Wistar , Analgesics/pharmacology , Neuralgia/drug therapy , Dose-Response Relationship, Drug
3.
J Pineal Res ; 73(4): e12825, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35996205

ABSTRACT

Melatonin, through its G protein-coupled receptor (GPCR) (MTNR1B gene) MT2 , is implicated in analgesia, but the relationship between MT2 receptors and the opioid system remains elusive. In a model of rodent neuropathic pain (spared nerve injured [SNI]), the selective melatonin MT2 agonist UCM924 reversed the allodynia (a pain response to a non-noxious stimulus), and this effect was nullified by the pharmacological blockade or genetic inactivation of the mu opioid receptor (MOR), but not the delta opioid receptor (DOR). Indeed, SNI MOR, but not DOR knockout mice, did not respond to the antiallodynic effects of the UCM924. Similarly, the nonselective opioid antagonist naloxone and the selective MOR antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) blocked the effects of UCM924 in SNI rats, but not the DOR antagonist naltrindole (NTI). Electrophysiological recordings in the rostral-ventromedial medulla (RVM) revealed that the typical reduction of the firing activity of pronociceptive ON-cells, and the enhancement of the firing of the antinociceptive OFF-cells, induced by the microinjection of the MT2 agonist UCM924 into the ventrolateral periaqueductal gray (vlPAG) were blocked by MOR, but not DOR, antagonism. Immunohistochemistry studies showed that MT2 receptors are expressed in both excitatory (CaMKIIα+ ) and inhibitory (GAD65+ ) neuronal cell bodies in the vlPAG (~2.16% total), but not RVM. Only 0.20% of vlPAG neurons coexpressed MOR and MT2 receptors. Finally, UCM924 treatment induced an increase in the enkephalin precursor gene (PENK) in the PAG of SNI mice. Collectively, the melatonin MT2 receptor agonism requires MORs to exert its antiallodynic effects, mostly through an interneuronal circuit involving MOR and MT2 receptors.


Subject(s)
Melatonin , Neuralgia , Mice , Animals , Rats , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/agonists , Melatonin/pharmacology , Melatonin/therapeutic use , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Receptors, Opioid, delta , Analgesics, Opioid/therapeutic use , Enkephalins/pharmacology , Enkephalins/therapeutic use , Naloxone/pharmacology , Naloxone/therapeutic use , Neuralgia/drug therapy
4.
Neuropsychopharmacology ; 47(6): 1188-1198, 2022 05.
Article in English | MEDLINE | ID: mdl-35301424

ABSTRACT

Lysergic acid diethylamide (LSD) is a serotonergic psychedelic compound receiving increasing interest due to putative anxiolytic and antidepressant properties. However, the potential neurobiological mechanisms mediating these effects remain elusive. Employing in vivo electrophysiology, microionthophoresis, behavioral paradigms and morphology assays, we assessed the impact of acute and chronic LSD administration on anxiety-like behavior, on the cortical dendritic spines and on the activity of serotonin (5-HT) neurons originating in the dorsal raphe nucleus (DRN) in male mice exposed to chronic restraint stress. We found that while the acute intraperitoneal (i.p.) administration of LSD (5, 15 and 30 and 60 µg/kg) did not produce any anxiolytic or antidepressant effects in non-stressed mice, the dose of 30 µg/kg (daily for 7 days) prevented the stress-induced anxiety-like behavior and the stress-induced decrease of cortical spine densitiy. Interestingly, while LSD acutely decreased the firing activity of 5-HT neurons, repeated LSD increased their basal firing rate and restored the low 5-HT firing induced by stress. This effect was accompanied by a decreased inhibitory response of 5-HT neurons to microiontophoretic applications of the 5-HT1A agonist 8-OH-DPAT (8-hydroxy-N,N-dipropyl-2-aminotetralin). In conclusion, repeated LSD prevents the exacerbation of anxiety-like behavior following chronic stress exposure, but has no behavioral effects in non-stressed mice. These effects are paralleled by increased cortical spinogenesis and an enhancement of 5-HT neurotransmission which might be due to 5-HT1A receptors desensitization. Increased cortical spine density and enhancement of serotonergic neurotransmission may thus represent a candidate mechanism which mediate the therapeutic effects of serotonergic psychedelics on stress-induced anxiety.


Subject(s)
Anti-Anxiety Agents , Hallucinogens , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety/drug therapy , Anxiety/etiology , Hallucinogens/pharmacology , Lysergic Acid Diethylamide/pharmacology , Male , Mice , Serotonin/pharmacology , Synaptic Transmission
5.
J Psychopharmacol ; 35(4): 469-482, 2021 04.
Article in English | MEDLINE | ID: mdl-33645311

ABSTRACT

BACKGROUND: The reticular thalamus gates thalamocortical information flow via finely tuned inhibition of thalamocortical cells in the mediodorsal thalamus. Brain imaging studies in humans show that the psychedelic lysergic acid diethylamide (LSD) modulates activity and connectivity within the cortico-striato-thalamo-cortical (CSTC) circuit, altering consciousness. However, the electrophysiological effects of LSD on the neurons in these brain areas remain elusive. METHODS: We employed in vivo extracellular single-unit recordings in anesthetized adult male mice to investigate the dose-response effects of cumulative LSD doses (5-160 µg/kg, intraperitoneal) upon reticular thalamus GABAergic neurons, thalamocortical relay neurons of the mediodorsal thalamus, and pyramidal neurons of the infralimbic prefrontal cortex. RESULTS: LSD decreased spontaneous firing and burst-firing activity in 50% of the recorded reticular thalamus neurons in a dose-response fashion starting at 10 µg/kg. Another population of neurons (50%) increased firing and burst-firing activity starting at 40 µg/kg. This modulation was accompanied by an increase in firing and burst-firing activity of thalamocortical neurons in the mediodorsal thalamus. On the contrary, LSD excited infralimbic prefrontal cortex pyramidal neurons only at the highest dose tested (160 µg/kg). The dopamine D2 receptor (D2) antagonist haloperidol administered after LSD increased burst-firing activity in the reticular thalamus neurons inhibited by LSD, decreased firing and burst-firing activity in the mediodorsal thalamus, and showed a trend towards further increasing the firing activity of neurons of the infralimbic prefrontal cortex. CONCLUSION: LSD modulates firing and burst-firing activity of reticular thalamus neurons and disinhibits mediodorsal thalamus relay neurons at least partially in a D2-mediated fashion. These effects of LSD on thalamocortical gating could explain its consciousness-altering effects in humans.


Subject(s)
Dose-Response Relationship, Drug , Electrophysiological Phenomena , Lysergic Acid Diethylamide/pharmacology , Prefrontal Cortex , Thalamus , Animals , Consciousness Disorders/chemically induced , Consciousness Disorders/metabolism , Dopamine D2 Receptor Antagonists/pharmacology , Hallucinogens/pharmacology , Male , Mice , Neural Pathways/drug effects , Neural Pathways/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Dopamine D2/metabolism , Thalamus/drug effects , Thalamus/metabolism
6.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article in English | MEDLINE | ID: mdl-33495318

ABSTRACT

Clinical studies have reported that the psychedelic lysergic acid diethylamide (LSD) enhances empathy and social behavior (SB) in humans, but its mechanism of action remains elusive. Using a multidisciplinary approach including in vivo electrophysiology, optogenetics, behavioral paradigms, and molecular biology, the effects of LSD on SB and glutamatergic neurotransmission in the medial prefrontal cortex (mPFC) were studied in male mice. Acute LSD (30 µg/kg) injection failed to increase SB. However, repeated LSD (30 µg/kg, once a day, for 7 days) administration promotes SB, without eliciting antidepressant/anxiolytic-like effects. Optogenetic inhibition of mPFC excitatory neurons dramatically inhibits social interaction and nullifies the prosocial effect of LSD. LSD potentiates the α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) and 5-HT2A, but not N-methyl-D-aspartate (NMDA) and 5-HT1A, synaptic responses in the mPFC and increases the phosphorylation of the serine-threonine protein kinases Akt and mTOR. In conditional knockout mice lacking Raptor (one of the structural components of the mTORC1 complex) in excitatory glutamatergic neurons (Raptorf/f:Camk2alpha-Cre), the prosocial effects of LSD and the potentiation of 5-HT2A/AMPA synaptic responses were nullified, demonstrating that LSD requires the integrity of mTORC1 in excitatory neurons to promote SB. Conversely, in knockout mice lacking Raptor in GABAergic neurons of the mPFC (Raptorf/f:Gad2-Cre), LSD promotes SB. These results indicate that LSD selectively enhances SB by potentiating mPFC excitatory transmission through 5-HT2A/AMPA receptors and mTOR signaling. The activation of 5-HT2A/AMPA/mTORC1 in the mPFC by psychedelic drugs should be explored for the treatment of mental diseases with SB impairments such as autism spectrum disorder and social anxiety disorder.


Subject(s)
Behavior, Animal/drug effects , Lysergic Acid Diethylamide/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Social Behavior , Synaptic Transmission/drug effects , Animals , Avoidance Learning/drug effects , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Optogenetics , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, AMPA/agonists , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Serotonin/metabolism , Synapses/drug effects , Synapses/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Nature ; 590(7845): 315-319, 2021 02.
Article in English | MEDLINE | ID: mdl-33328636

ABSTRACT

Effective pharmacotherapy for major depressive disorder remains a major challenge, as more than 30% of patients are resistant to the first line of treatment (selective serotonin reuptake inhibitors)1. Sub-anaesthetic doses of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist2,3, provide rapid and long-lasting antidepressant effects in these patients4-6, but the molecular mechanism of these effects remains unclear7,8. Ketamine has been proposed to exert its antidepressant effects through its metabolite (2R,6R)-hydroxynorketamine ((2R,6R)-HNK)9. The antidepressant effects of ketamine and (2R,6R)-HNK in rodents require activation of the mTORC1 kinase10,11. mTORC1 controls various neuronal functions12, particularly through cap-dependent initiation of mRNA translation via the phosphorylation and inactivation of eukaryotic initiation factor 4E-binding proteins (4E-BPs)13. Here we show that 4E-BP1 and 4E-BP2 are key effectors of the antidepressant activity of ketamine and (2R,6R)-HNK, and that ketamine-induced hippocampal synaptic plasticity depends on 4E-BP2 and, to a lesser extent, 4E-BP1. It has been hypothesized that ketamine activates mTORC1-4E-BP signalling in pyramidal excitatory cells of the cortex8,14. To test this hypothesis, we studied the behavioural response to ketamine and (2R,6R)-HNK in mice lacking 4E-BPs in either excitatory or inhibitory neurons. The antidepressant activity of the drugs is mediated by 4E-BP2 in excitatory neurons, and 4E-BP1 and 4E-BP2 in inhibitory neurons. Notably, genetic deletion of 4E-BP2 in inhibitory neurons induced a reduction in baseline immobility in the forced swim test, mimicking an antidepressant effect. Deletion of 4E-BP2 specifically in inhibitory neurons also prevented the ketamine-induced increase in hippocampal excitatory neurotransmission, and this effect concurred with the inability of ketamine to induce a long-lasting decrease in inhibitory neurotransmission. Overall, our data show that 4E-BPs are central to the antidepressant activity of ketamine.


Subject(s)
Antidepressive Agents/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Ketamine/pharmacology , Neurons/drug effects , Neurons/metabolism , Protein Biosynthesis/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Depressive Disorder, Major/drug therapy , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Interneurons/metabolism , Ketamine/analogs & derivatives , Ketamine/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mutation , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/classification , Neurons/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synaptic Transmission/drug effects
8.
J Pineal Res ; 69(3): e12671, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32430930

ABSTRACT

Melatonin, a neurohormone that binds to two G protein-coupled receptors MT1 and MT2, is involved in pain regulation, but the distinct role of each receptor has yet to be defined. We characterized the nociceptive responses of mice with genetic inactivation of melatonin MT1 (MT1 -/- ), or MT2 (MT2 -/- ), or both MT1 /MT2 (MT1 -/- /MT2 -/- ) receptors in the hot plate test (HPT), and the formalin test (FT). In HPT and FT, MT1 -/- display no differences compared to their wild-type littermates (CTL), whereas both MT2 -/- and MT1 -/- /MT2 -/- mice showed a reduced thermal sensitivity and a decreased tonic nocifensive behavior during phase 2 of the FT in the light phase. The MT2 partial agonist UCM924 induced an antinociceptive effect in MT1 -/- but not in MT2 -/- and MT1 -/- /MT2 -/- mice. Also, the competitive opioid antagonist naloxone had no effects in CTL, whereas it induced a decrease of nociceptive thresholds in MT2 -/- mice. Our results show that the genetic inactivation of melatonin MT2 , but not MT1 receptors, produces a distinct effect on nociceptive threshold, suggesting that the melatonin MT2 receptor subtype is selectively involved in the regulation of pain responses.


Subject(s)
Melatonin , Nociception , Receptor, Melatonin, MT1 , Receptor, Melatonin, MT2 , Animals , Melatonin/genetics , Melatonin/metabolism , Mice , Mice, Knockout , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/metabolism
9.
Int J Mol Sci ; 20(10)2019 May 17.
Article in English | MEDLINE | ID: mdl-31108968

ABSTRACT

Melatonin (MLT) is a neurohormone that regulates many physiological functions including sleep, pain, thermoregulation, and circadian rhythms. MLT acts mainly through two G-protein-coupled receptors named MT1 and MT2, but also through an MLT type-3 receptor (MT3). However, the role of MLT receptor subtypes in thermoregulation is still unknown. We have thus investigated the effects of selective and non-selective MLT receptor agonists/antagonists on body temperature (Tb) in rats across the 12/12-h light-dark cycle. Rectal temperature was measured every 15 min from 4:00 a.m. to 9:30 a.m. and from 4:00 p.m. to 9:30 p.m., following subcutaneous injection of each compound at either 5:00 a.m. or 5:00 p.m. MLT (40 mg/kg) had no effect when injected at 5 a.m., whereas it decreased Tb during the light phase only when injected at 5:00 p.m. This effect was blocked by the selective MT2 receptor antagonist 4P-PDOT and the non-selective MT1/MT2 receptor antagonist, luzindole, but not by the α1/MT3 receptors antagonist prazosin. However, unlike MLT, neither the selective MT1 receptor partial agonist UCM871 (14 mg/kg) nor the selective MT2 partial agonist UCM924 (40 mg/kg) altered Tb during the light phase. In contrast, UCM871 injected at 5:00 p.m. increased Tb at the beginning of the dark phase, whereas UCM924 injected at 5:00 a.m. decreased Tb at the end of the dark phase. These effects were blocked by luzindole and 4P-PDOT, respectively. The MT3 receptor agonist GR135531 (10 mg/kg) did not affect Tb. These data suggest that the simultaneous activation of both MT1 and MT2 receptors is necessary to regulate Tb during the light phase, whereas in a complex but yet unknown manner, they regulate Tb differently during the dark phase. Overall, MT1 and MT2 receptors display complementary but also distinct roles in modulating circadian fluctuations of Tb.


Subject(s)
Body Temperature/drug effects , Melatonin/administration & dosage , Receptor, Melatonin, MT1/agonists , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/agonists , Acetamides/administration & dosage , Acetamides/pharmacology , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Injections, Subcutaneous , Male , Melatonin/pharmacology , Photoperiod , Rats , Rats, Wistar , Receptor, Melatonin, MT1/antagonists & inhibitors , Receptor, Melatonin, MT2/antagonists & inhibitors , Receptor, Melatonin, MT2/metabolism , Tetrahydronaphthalenes/administration & dosage , Tetrahydronaphthalenes/pharmacology , Tryptamines/administration & dosage , Tryptamines/pharmacology
10.
Pharmacol Res ; 144: 343-356, 2019 06.
Article in English | MEDLINE | ID: mdl-31029764

ABSTRACT

Melatonin (MLT), a neuromodulator mainly acting through two G-protein coupled receptors MT1 and MT2, regulates many brain functions, including circadian rhythms, mood, pain and sleep. MLT and non-selective MT1/MT2 receptor agonists are clinically used in neuropsychiatric and/or sleep disorders. However, the selective roles of the MT1 and MT2 receptors need to be clarified. Here, we review the role of the MT1 receptor in neuropsychopharmacology, describe the anatomical localization of MT1 receptors in the brain, discuss the medicinal chemistry, biochemistry and molecular aspects of the receptor, and explore the findings linking MT1 receptors to psychiatric and neurological disorders. MT1 receptors are localized in brain regions which regulate circadian rhythms, sleep, and mood, such as the suprachiasmatic nucleus, cortex, hippocampus, dorsal raphe nucleus and lateral hypothalamus. Their activation modulates intracellular signaling pathways also targeted by psychoactive drugs, including antidepressants and mood stabilizers. MT1 receptor knockout mice display increased anxiety, a depressive-like phenotype, increased propensity to reward and addiction, and reduced Rapid-Eye-Movement sleep. These behavioral dysfunctions are associated with altered serotonergic and noradrenergic neurotransmissions. Several studies indicate that the MT1, rather than MT2, receptor is implicated in circadian rhythm regulation. The involvement of MT1 receptors in Alzheimer's and Huntington diseases has also been proposed. Postmortem studies in depressed patients have further confirmed the possible involvement of MT1 receptors in depression. Overall, there is substantial evidence indicating a role for MT1 receptor in modulating brain function and mood. Consequently, this MLT receptor subtype deserves to be further examined as a novel target for neuropsychopharmacological drug development.


Subject(s)
Receptor, Melatonin, MT1/metabolism , Animals , Circadian Rhythm/drug effects , Drug Discovery , Humans , Ligands , Molecular Targeted Therapy , Mood Disorders/drug therapy , Mood Disorders/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Psychotic Disorders/drug therapy , Psychotic Disorders/metabolism , Receptor, Melatonin, MT1/analysis , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/metabolism , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism
11.
Pain ; 160(1): 136-150, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30157131

ABSTRACT

Clinical studies indicate that cannabidiol (CBD), the primary nonaddictive component of cannabis that interacts with the serotonin (5-HT)1A receptor, may possess analgesic and anxiolytic effects. However, its effects on 5-HT neuronal activity, as well as its impact on models of neuropathic pain are unknown. First, using in vivo single-unit extracellular recordings in rats, we demonstrated that acute intravenous (i.v.) increasing doses of CBD (0.1-1.0 mg/kg) decreased the firing rate of 5-HT neurons in the dorsal raphe nucleus, which was prevented by administration of the 5-HT1A antagonist WAY 100635 (0.3 mg/kg, i.v.) and the TRPV1 antagonist capsazepine (1 mg/kg, i.v.) but not by the CB1 receptor antagonist AM 251 (1 mg/kg, i.v.). Repeated treatment with CBD (5 mg/kg/day, subcutaneously [s.c.], for 7 days) increased 5-HT firing through desensitization of 5-HT1A receptors. Rats subjected to the spared nerve injury model for 24 days showed decreased 5-HT firing activity, mechanical allodynia, and increased anxiety-like behavior in the elevated plus maze test, open-field test, and novelty-suppressed feeding test. Seven days of treatment with CBD reduced mechanical allodynia, decreased anxiety-like behavior, and normalized 5-HT activity. Antiallodynic effects of CBD were fully prevented by capsazepine (10 mg/kg/day, s.c., for 7 days) and partially prevented by WAY 100635 (2 mg/kg/day, s.c., for 7 days), whereas the anxiolytic effect was blocked only by WAY. Overall, repeated treatment with low-dose CBD induces analgesia predominantly through TRPV1 activation, reduces anxiety through 5-HT1A receptor activation, and rescues impaired 5-HT neurotransmission under neuropathic pain conditions.


Subject(s)
Anxiety/drug therapy , Anxiety/etiology , Cannabidiol/therapeutic use , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Neuralgia/complications , Serotonin/metabolism , Action Potentials/drug effects , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Disease Models, Animal , Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Ganglia, Spinal/cytology , Hyperalgesia/therapy , Lysergic Acid Diethylamide/pharmacology , Male , Maze Learning/drug effects , Neuralgia/pathology , Piperazines/therapeutic use , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyridines/therapeutic use , Rats , Rats, Wistar , Serotonin Antagonists/pharmacology , Swimming
12.
Brain Res Bull ; 146: 40-50, 2019 03.
Article in English | MEDLINE | ID: mdl-30593876

ABSTRACT

Behavioral flexibility is a complex cognitive function that allows for the rapid adaptation to a changing environment. This ability is modulated by the proper function of the prefrontal cortex (PFC), which receives important projections from the ventral hippocampus (vHPC). In this context, the vHPC might play a very important role in behavioral flexibility. Here, we infused the voltage-gated sodium channel blocker tetrodotoxin (TTX) to bilaterally inactivate the vHPC in adult rats and assessed behavioral flexibility in a spatial setting, using the allocentric-egocentric strategy switching task in the cross-shaped maze. We demonstrate that bilateral inactivation of the vHPC impaired the ability to switch from allocentric to egocentric (Experiment 1), and from egocentric to allocentric (Experiment 2) spatial strategies, as noted by the increased number of trials to reach the learning criterion and of entries into incorrect arms. These results resembled the effects of PFC inactivation by TTX on behavioral flexibility (Experiment 3). Furthermore, TTX infusion in the vHPC did not affect allocentric or egocentric learning per se but the ability to switch between either spatial strategy. Remarkably, inactivation of the vHPC decreased the latency to select an arm during the transition from an allocentric to an egocentric strategy, suggesting that the vHPC might mediate impulsive choices during the acquisition of a novel task. Our results highlight an important role of the vHPC in mediating behavioral flexibility by, most likely, modulating proper PFC function.


Subject(s)
Behavior, Animal/physiology , Hippocampus/physiology , Learning/physiology , Altruism , Animals , Cognition/physiology , Egocentrism , Hippocampus/metabolism , Male , Maze Learning/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Social Behavior , Space Perception/physiology , Tetrodotoxin/pharmacology
13.
Eur J Pharmacol ; 764: 424-432, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26162699

ABSTRACT

Melatonin is a neurohormone involved in the regulation of both acute and chronic pain whose mechanism is still not completely understood. We have recently demonstrated that selective MT2 melatonin receptor partial agonists have antiallodynic properties in animal models of chronic neuropathic pain by modulating ON/OFF cells of the descending antinociceptive system. Here, we examined the antinociceptive properties of the selective MT2 melatonin receptor partial agonists N-{2-[(3-methoxyphenyl)phenylamino]ethyl}acetamide (UCM765) and N-{2-[(3-bromophenyl)-(4-fluorophenyl)amino]ethyl}acetamide (UCM924) in two animal models of acute and inflammatory pain: the hot-plate and formalin tests. UCM765 and UCM924 (5-40 mg/kg, s.c.) dose-dependently increased the temperature of the first hind paw lick in the hot-plate test, and decreased the total time spent licking the injected hind paw in the formalin test. Antinociceptive effects of UCM765 and UCM924 were maximal at the dose of 20mg/kg. At this dose, the effects of UCM765 and UCM924 were similar to those produced by 200 mg/kg acetaminophen in the hot-plate test, and by 3 mg/kg ketorolac or 150 mg/kg MLT in the formalin test. Notably, antinociceptive effects of the two MT2 partial agonists were blocked by the pre-treatment with the MT2 antagonist 4-phenyl-2-propionamidotetralin (4P-PDOT, 10 mg/kg) in both paradigms. These results demonstrate the antinociceptive properties of UCM765 and UCM924 in acute and inflammatory pain models and corroborate the concept that MT2 melatonin receptor may be a novel target for analgesic drug development.


Subject(s)
Acetamides/pharmacology , Analgesics/pharmacology , Aniline Compounds/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Nociception/drug effects , Nociceptive Pain/prevention & control , Receptor, Melatonin, MT2/agonists , Acetamides/metabolism , Acetaminophen/pharmacology , Analgesics/metabolism , Aniline Compounds/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Partial Agonism , Formaldehyde , Ketorolac/pharmacology , Male , Nociceptive Pain/chemically induced , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Protein Binding , Rats, Wistar , Receptor, Melatonin, MT2/metabolism , Signal Transduction/drug effects , Tetrahydronaphthalenes/pharmacology , Time Factors
14.
Pain ; 156(2): 305-317, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25599452

ABSTRACT

Neuropathic pain is an important public health problem for which only a few treatments are available. Preclinical studies show that melatonin (MLT), a neurohormone acting on MT1 and MT2 receptors, has analgesic properties, likely through MT2 receptors. Here, we determined the effects of the novel selective MLT MT2 receptor partial agonist N-{2-([3-bromophenyl]-4-fluorophenylamino)ethyl}acetamide (UCM924) in 2 neuropathic pain models in rats and examined its supraspinal mechanism of action. In rat L5-L6 spinal nerve ligation and spared nerve injury models, UCM924 (20-40 mg/kg, subcutaneously) produced a prolonged antinociceptive effect that is : (1) dose-dependent and blocked by the selective MT2 receptor antagonist 4-phenyl-2-propionamidotetralin, (2) superior to a high dose of MLT (150 mg/kg) and comparable with gabapentin (100 mg/kg), but (3) without noticeable motor coordination impairments in the rotarod test. Using double staining immunohistochemistry, we found that MT2 receptors are expressed by glutamatergic neurons in the rostral ventrolateral periaqueductal gray. Using in vivo electrophysiology combined with tail flick, we observed that microinjection of UCM924 into the ventrolateral periaqueductal gray decreased tail flick responses, depressed the firing activity of ON cells, and activated the firing of OFF cells; all effects were MT2 receptor-dependent. Altogether, these data demonstrate that selective MT2 receptor partial agonists have analgesic properties through modulation of brainstem descending antinociceptive pathways, and MT2 receptors may represent a novel target in the treatment of neuropathic pain.


Subject(s)
Acetamides/metabolism , Aniline Compounds/metabolism , Brain Stem/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Pyramidal Tracts/metabolism , Receptor, Melatonin, MT2/metabolism , Acetamides/pharmacology , Acetamides/therapeutic use , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Brain Stem/drug effects , Ligands , Male , Pain Measurement/drug effects , Pain Measurement/methods , Pyramidal Tracts/drug effects , Rats , Rats, Wistar , Receptor, Melatonin, MT2/agonists
15.
Synapse ; 68(10): 454-67, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24955825

ABSTRACT

Direct evidence that dopamine (DA) neurotransmission varies during the 24 h of the day is lacking. Here, we have characterized the firing activity of DA neurons located in the ventral tegmental area (VTA) using single-unit extracellular recordings in anesthetized rats kept on a standard light-dark cycle. DA neuronal firing activity was measured under basal conditions and in response to intravenous administration of increasing doses of amphetamine (AMPH: 0.5, 1, 2, 5 mg/kg), apomorphine (APO: 25, 50, 100, 200 µg/kg) and melatonin (MLT: 0.1, 1, 10 mg/kg) at different time intervals of the light-dark cycle. DA firing activity peaked between 07:00 and 11:00 h (3.5 ± 0.3 Hz) and between 19:00 and 23:00 h (4.1 ± 0.7 Hz), with lowest activity occurring between 11:00 and 15:00 h (2.4 ± 0.2 Hz) and between 23:00 and 03:00 h (2.6 ± 0.2 Hz). The highest number of spontaneously active neurons was observed between 03:00 and 06:00 h (2.5 ± 0.3 neurons/track), whereas the lowest was between 19:00 and 23:00 h (1.5 ± 0.2 neurons/track). The inhibitory effect of AMPH on DA firing rate was similar in both phases. The inhibitory effect of low dose of APO (25 µg/kg, dose selective for D2 autoreceptor) was more potent in the dark phase, whereas APO effects at higher doses were similar in both phases. Finally, MLT administration (1 mg/kg) produced a moderate inhibition of DA cell firing in both phases. These experiments demonstrate the existence of an intradiurnal rhythmic pattern of VTA DA neuronal firing activity and a higher pharmacological response of D2 autoreceptors in the dark phase.


Subject(s)
Circadian Rhythm/physiology , Dopaminergic Neurons/physiology , Ventral Tegmental Area/physiology , Action Potentials/drug effects , Amphetamine/pharmacology , Animals , Apomorphine/pharmacology , Central Nervous System Depressants/pharmacology , Circadian Rhythm/drug effects , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Male , Melatonin/pharmacology , Microelectrodes , Photoperiod , Rats, Sprague-Dawley , Ventral Tegmental Area/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...