Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Methods Mol Biol ; 2550: 391-411, 2022.
Article in English | MEDLINE | ID: mdl-36180708

ABSTRACT

The neurohormone melatonin facilitates entrainment of biological rhythms to environmental light-dark conditions as well as phase-shifts of circadian rhythms in constant conditions via activation of the MT1 and/or MT2 receptors expressed within the suprachiasmatic nucleus of the hypothalamus. The efficacy of melatonin and related agonists to modulate biological rhythms can be assessed using two well-validated mouse models of rhythmic behaviors. These models serve as predictive measures of therapeutic efficacy for treatment of circadian phase disorders caused by internal (e.g., clock gene mutations, blindness, depression, seasonal affective disorder) or external (e.g., shift work, travel across time zones) causes in humans. Here we provide background and detailed protocols for quantitative assessment of the magnitude and efficacy of melatonin receptor ligands in mouse circadian phase-shift and re-entrainment paradigms. The utility of these models in the discovery of novel therapeutics acting on melatonin receptors will also be discussed.


Subject(s)
Melatonin , Animals , Circadian Rhythm/genetics , Disease Models, Animal , Humans , Ligands , Melatonin/pharmacology , Melatonin/therapeutic use , Mice , Receptors, Melatonin/agonists , Receptors, Melatonin/genetics , Receptors, Melatonin/metabolism , Suprachiasmatic Nucleus/metabolism
2.
Molecules ; 27(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36080418

ABSTRACT

Melatonin (MEL), an indolamine with diverse functions in the brain, has been shown to produce antidepressant-like effects, presumably through stimulating neurogenesis. We recently showed that the combination of MEL with ketamine (KET), an NMDA receptor antagonist, has robust antidepressant-like effects in mice, at doses that, by themselves, are non-effective and have no adverse effects. Here, we show that the KET/MEL combination increases neurogenesis in a clone derived from human olfactory neuronal precursors, a translational pre-clinical model for effects in the human CNS. Neurogenesis was assessed by the formation of cell clusters > 50 µm in diameter, positively stained for nestin, doublecortin, BrdU and Ki67, markers of progenitor cells, neurogenesis, and proliferation. FGF, EGF and BDNF growth factors increased the number of cell clusters in cultured, cloned ONPs. Similarly, KET or MEL increased the number of clusters in a dose-dependent manner. The KET/MEL combination further increased the formation of clusters, with a maximal effect obtained after a triple administration schedule. Our results show that the combination of KET/MEL, at subeffective doses that do not produce adverse effects, stimulate neurogenesis in human neuronal precursors. Moreover, the mechanism by which the combination elicits neurogenesis is meditated by melatonin receptors, CaM Kinase II and CaM antagonism. This could have clinical advantages for the fast treatment of depression.


Subject(s)
Ketamine , Melatonin , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Hippocampus/metabolism , Humans , Ketamine/metabolism , Ketamine/pharmacology , Melatonin/metabolism , Melatonin/pharmacology , Mice , Neurogenesis , Neurons
3.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34502152

ABSTRACT

Major depressive disorder is a disabling disease with the number of affected individuals increasing each year. Current antidepressant treatments take between three to six weeks to be effective with forty percent of patients being resistant to treatment, making it necessary to search for new antidepressant treatments. Ketamine, a phencyclidine hydrochloride derivative, given intravenously, induces a rapid antidepressant effect in humans. In mice, it causes increased neurogenesis and antidepressant-like effects. However, it also produces psychomimetic effects in humans and in rodents increases the locomotor activity. In contrast, melatonin, a hormone secreted by the pineal gland and synthesized in extrapineal sites, increases new neuron formation and causes antidepressant-like effects in adult rodents with no collateral effects. Here, we assessed the effects of a non-effective dose of ketamine in combination with melatonin (KET/MEL), both on neurogenesis as well as on the antidepressant-like effect in mice. Our results showed that KET/MEL combination increased neurogenesis and produced antidepressant-like effects without altering locomotor activity after both single and triple administration protocols. Our data strongly suggest that KET/MEL combination could be used to simultaneously promote neurogenesis, reverting neuronal atrophy and inducing antidepressant-like effects.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/drug therapy , Ketamine/therapeutic use , Melatonin/therapeutic use , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacology , Drug Combinations , Drug Synergism , Ketamine/administration & dosage , Ketamine/pharmacology , Male , Melatonin/administration & dosage , Melatonin/pharmacology , Mice , Neurogenesis/drug effects
4.
J Pineal Res ; 71(2): e12748, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34085306

ABSTRACT

The hormone melatonin is synthesized from serotonin by two enzymatic reactions (AANAT and ASMT/HIOMT) in the pineal gland following a circadian rhythm with low levels during the day and high levels at night. The robust nightly peak of melatonin secretion is an output signal of the circadian clock to the whole organism. However, so far the regulatory roles of endogenous melatonin in mammalian biological rhythms and physiology processes are poorly understood. Here, we establish congenic mouse lines (>N10 generations) that are proficient or deficient in melatonin synthesis (AH+/+ or AH-/- mice, respectively) on the C57BL/6J genetic background by crossing melatonin-proficient MSM/Ms with C57BL/6J. AH+/+ mice displayed robust nightly peak of melatonin secretion and had significantly higher levels of pineal and plasma melatonin vs AH-/- mice. Using this mice model, we investigated the role of endogenous melatonin in regulating multiple biological rhythms, physiological processes, and rhythmic behaviors. In the melatonin-proficient (AH+/+) mice, the rate of re-entrainment of wheel-running activity was accelerated following a 6-hour phase advance of dark onset when comparted with AH-/- mice, suggesting a role of endogenous melatonin in facilitating clock adjustment. Further in the AH+/+ mice, there was a significant decrease in body weight, gonadal weight and reproductive performance, and a significant increase in daily torpor (a hypothermic and hypometabolic state lasting only hours during adverse conditions). Endogenous melatonin, however, had no effect in the modulation of the diurnal rhythm of 2-[125 I]-iodomelatonin receptor expression in the SCN, free-running wheel behavior in constant darkness, life span, spontaneous homecage behaviors, and various types of social-emotional behaviors. The findings also shed light on the role of endogenous melatonin in mice domestication and provide new insights into melatonin's action in reducing energy expenditure during a food shortage. In summary, the congenic mice model generated in this study offers a significant advantage toward understanding of the role of endogenous melatonin in regulating melatonin receptor-mediated rhythm behaviors and physiological functions.


Subject(s)
Melatonin , Pineal Gland , Animals , Circadian Rhythm/physiology , Melatonin/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Pineal Gland/metabolism , Reproduction
5.
J Pineal Res ; 71(1): e12732, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33759236

ABSTRACT

Data indicate that controlling inflammatory responses to COVID-19 may be as important as antiviral therapies or could be an important adjunctive approach. Melatonin possesses anti-inflammation, antioxidation, and immune-enhancing features directly and/or indirectly through its own receptor signaling and is therefore well suited to reduce the severity of COVID-19. Studies have proposed that melatonin regulates COVID-19-associated proteins directly through regulation of molecules such as calmodulin (CALM) 1 and CALM 2, calreticulin (CalR), or myeloperoxidase (MPO) and/or indirectly through actions on GPCR (eg, MTNR1A, MTNR1B) and nuclear (eg, RORα, RORß) melatonin receptor signaling. However, the exact mechanism(s) and doses by which melatonin reduces the severity of COVID-19 is still open for debate, warranting the need for further testing of melatonin in placebo-controlled randomized clinical trials for COVID-19.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19 Drug Treatment , Melatonin/therapeutic use , Receptors, Melatonin/agonists , SARS-CoV-2/pathogenicity , COVID-19/immunology , COVID-19/metabolism , COVID-19/virology , Host-Pathogen Interactions , Humans , Receptors, Melatonin/metabolism , SARS-CoV-2/immunology , Severity of Illness Index , Signal Transduction
6.
J Pharmacol Exp Ther ; 376(2): 306-321, 2021 02.
Article in English | MEDLINE | ID: mdl-33203660

ABSTRACT

Integrated in silico chemical clustering and melatonin receptor molecular modeling combined with in vitro 2-[125I]-iodomelatonin competition binding were used to identify carbamate insecticides with affinity for human melatonin receptor 1 (hMT1) and human melatonin receptor 2 (hMT2). Saturation and kinetic binding studies with 2-[125I]-iodomelatonin revealed lead carbamates (carbaryl, fenobucarb, bendiocarb, carbofuran) to be orthosteric ligands with antagonist apparent efficacy at hMT1 and agonist apparent efficacy at hMT2 Furthermore, using quantitative receptor autoradiography in coronal brain slices from C3H/HeN mice, carbaryl, fenobucarb, and bendiocarb competed for 2-[125I]-iodomelatonin binding in the suprachiasmatic nucleus (SCN), paraventricular nucleus of the thalamus (PVT), and pars tuberalis (PT) with affinities similar to those determined for the hMT1 receptor. Carbaryl (10 mg/kg i.p.) administered in vivo also competed ex vivo for 2-[125I]-iodomelatonin binding to the SCN, PVT, and PT, demonstrating the ability to reach brain melatonin receptors in C3H/HeN mice. Furthermore, the same dose of carbaryl given to C3H/HeN mice in constant dark for three consecutive days at subjective dusk (circadian time 10) phase-advanced circadian activity rhythms (mean = 0.91 hours) similar to melatonin (mean = 1.12 hours) when compared with vehicle (mean = 0.04 hours). Carbaryl-mediated phase shift of overt circadian activity rhythm onset is likely mediated via interactions with SCN melatonin receptors. Based on the pharmacological actions of carbaryl and other carbamate insecticides at melatonin receptors, exposure may modulate time-of-day information conveyed to the master biologic clock relevant to adverse health outcomes. SIGNIFICANCE STATEMENT: In silico chemical clustering and molecular modeling in conjunction with in vitro bioassays identified several carbamate insecticides (i.e., carbaryl, carbofuran, fenobucarb, bendiocarb) as pharmacologically active orthosteric melatonin receptor 1 and 2 ligands. This work further demonstrated that carbaryl competes for melatonin receptor binding in the master biological clock (suprachiasmatic nucleus) and phase-advances overt circadian activity rhythms in C3H/HeN mice, supporting the relevance of circadian effects when interpreting toxicological findings related to carbamate insecticide exposure.


Subject(s)
Carbamates/pharmacology , Insecticides/pharmacology , Receptors, Melatonin/metabolism , Animals , Binding Sites , Brain/drug effects , Brain/metabolism , CHO Cells , Carbamates/pharmacokinetics , Circadian Rhythm/drug effects , Cricetinae , Cricetulus , Humans , Insecticides/pharmacokinetics , Male , Mice , Molecular Docking Simulation , Protein Binding , Receptors, Melatonin/chemistry
7.
Nature ; 579(7800): 609-614, 2020 03.
Article in English | MEDLINE | ID: mdl-32040955

ABSTRACT

The neuromodulator melatonin synchronizes circadian rhythms and related physiological functions through the actions of two G-protein-coupled receptors: MT1 and MT2. Circadian release of melatonin at night from the pineal gland activates melatonin receptors in the suprachiasmatic nucleus of the hypothalamus, synchronizing the physiology and behaviour of animals to the light-dark cycle1-4. The two receptors are established drug targets for aligning circadian phase to this cycle in disorders of sleep5,6 and depression1-4,7-9. Despite their importance, few in vivo active MT1-selective ligands have been reported2,8,10-12, hampering both the understanding of circadian biology and the development of targeted therapeutics. Here we docked more than 150 million virtual molecules to an MT1 crystal structure, prioritizing structural fit and chemical novelty. Of these compounds, 38 high-ranking molecules were synthesized and tested, revealing ligands with potencies ranging from 470 picomolar to 6 micromolar. Structure-based optimization led to two selective MT1 inverse agonists-which were topologically unrelated to previously explored chemotypes-that acted as inverse agonists in a mouse model of circadian re-entrainment. Notably, we found that these MT1-selective inverse agonists advanced the phase of the mouse circadian clock by 1.3-1.5 h when given at subjective dusk, an agonist-like effect that was eliminated in MT1- but not in MT2-knockout mice. This study illustrates the opportunities for modulating melatonin receptor biology through MT1-selective ligands and for the discovery of previously undescribed, in vivo active chemotypes from structure-based screens of diverse, ultralarge libraries.


Subject(s)
Circadian Rhythm/physiology , Ligands , Receptors, Melatonin/agonists , Receptors, Melatonin/metabolism , Animals , Circadian Rhythm/drug effects , Darkness , Drug Evaluation, Preclinical , Drug Inverse Agonism , Female , Humans , Light , Male , Mice , Mice, Knockout , Molecular Docking Simulation , Receptor, Melatonin, MT1/agonists , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/agonists , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Receptors, Melatonin/deficiency , Receptors, Melatonin/genetics , Small Molecule Libraries/pharmacology , Substrate Specificity/genetics
8.
Int J Mol Sci ; 19(8)2018 Aug 03.
Article in English | MEDLINE | ID: mdl-30081472

ABSTRACT

Melatonin is synthesized by the pineal gland with a circadian rhythm in synchrony with the environmental light/dark cycle. A gradual increase in circulating levels of melatonin occur after lights off, reaching its maximum around the middle of the dark phase. Agonists of melatonin receptors have proved effectiveness as antidepressants in clinical trials. However, there is contradictory evidence about the potential antidepressant effect of melatonin itself. Herein we studied melatonin administration in mice at two zeitgeber times (ZT; ZT = 0 lights on; 12:12 L/D), one hour before the beginning (ZT11) and at the middle (ZT18) of the dark phase after either a single or a three-dose protocol. Behavioral despair was assessed through a forced-swimming test (FST) or a tail suspension test (TST), at ZT18.5. A single dose of 4 mg/kg melatonin at ZT11 was effective to reduce the immobility time in both tests. However, acute administration of melatonin at ZT18 was not effective in mice subjected to FST, and a higher dose (16 mg/kg) was required to reduce immobility time in the TST. A three-dose administration protocol of 16 mg/kg melatonin (ZT18, ZT11, and ZT18) significantly reduced immobility time in FST. Data indicate that the timely administration of melatonin could improve its antidepressant-like effect.


Subject(s)
Antidepressive Agents/therapeutic use , Melatonin/therapeutic use , Animals , Antidepressive Agents/blood , Depression/drug therapy , Disease Models, Animal , Hindlimb Suspension , Male , Melatonin/blood , Mice , Swimming/physiology
9.
Br J Pharmacol ; 175(16): 3200-3208, 2018 08.
Article in English | MEDLINE | ID: mdl-29512136

ABSTRACT

Mood disorders are a spectrum of neuropsychiatric disorders characterized by changes in the emotional state. In particular, major depressive disorder is expected to have a worldwide prevalence of 20% in 2020, representing a huge socio-economic burden. Currently used antidepressant drugs have poor efficacy with only 30% of the patients in remission after the first line of treatment. Importantly, mood disorder patients present uncoupling of circadian rhythms. In this regard, melatonin (5-methoxy-N-acetyltryptamine), an indolamine synthesized by the pineal gland during the night, contributes to synchronization of body rhythms with the environmental light/dark cycle. In this review, we describe evidence supporting antidepressant-like actions of melatonin related to the circadian modulation of neuroplastic changes in the hippocampus. We also present evidence for the role of melatonin receptors and their signalling pathways underlying modulatory effects in neuroplasticity. Finally, we briefly discuss the detrimental consequences of circadian disruption on neuroplasticity and mood disorders, due to the modern human lifestyle. Together, data suggest that melatonin's stimulation of neurogenesis and neuronal differentiation is beneficial to patients with mood disorders. LINKED ARTICLES: This article is part of a themed section on Recent Developments in Research of Melatonin and its Potential Therapeutic Applications. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.16/issuetoc.


Subject(s)
Circadian Rhythm/physiology , Depression/drug therapy , Melatonin/physiology , Neuronal Plasticity/physiology , Animals , Depression/metabolism , Depression/physiopathology , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Melatonin/therapeutic use
10.
Behav Brain Res ; 343: 28-35, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29374562

ABSTRACT

Palatable food is known for its ability to enhance reinforcing responses. Studies have suggested a circadian variation in both drug and natural reinforcement, with each following its own time course. The goal of this study was to determine the role of the MT1 and MT2 melatonin receptors in palatable snack food-induced reinforcement, as measured by the conditioned place preference (CPP) paradigm during the light and dark phases. C3H/HeN wild-type mice were trained for snack food-induced CPP at either ZT 6 - 8 (ZT: Zeitgeber time; ZT 0 = lights on), when endogenous melatonin levels are low, or ZT 19 - 21, when melatonin levels are high. These time points also correspond to the high and low points for expression of the circadian gene Period1, respectively. The amount of snack food (chow, Cheetos®, Froot Loops® and Oreos®) consumed was of similar magnitude at both times, however only C3H/HeN mice conditioned to snack food at ZT 6 - 8 developed a place preference. C3H/HeN mice with a genetic deletion of either the MT1 (MT1KO) or MT2 (MT2KO) receptor tested at ZT 6 - 8 did not develop a place preference for snack food. Although the MT2KO mice showed a similar amount of snack food consumed when compared to wild-type mice, the MT1KO mice consumed significantly less than either genotype. We conclude that in our mouse model snack food-induced CPP is dependent on time of day and the presence of the MT1 or MT2 receptors, suggesting a role for melatonin and its receptors in snack food-induced reinforcement.


Subject(s)
Feeding Behavior/physiology , Feeding Behavior/psychology , Food , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , Reinforcement, Psychology , Animals , Conditioning, Psychological/physiology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Photoperiod , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Spatial Behavior/physiology
11.
J Clin Transl Sci ; 2(2): 95-102, 2018 Apr.
Article in English | MEDLINE | ID: mdl-31660222

ABSTRACT

INTRODUCTION: The Best Practices in Social and Behavioral Research Course was developed to provide instruction on good clinical practice for social and behavioral trials. This study evaluated the new course. METHODS: Participants across 4 universities took the course (n=294) and were sent surveys following course completion and 2 months later. Outcomes included relevance, how engaging the course was, and working differently because of the course. Open-ended questions were posed to understand how work was impacted. RESULTS: Participants rated the course as relevant and engaging (6.4 and 5.8/7 points) and reported working differently (4.7/7 points). Participants with less experience in social and behavioral trials were most likely to report working differently 2 months later. DISCUSSION: The course was perceived as relevant and engaging. Participants described actions taken to improve rigor in implementing trials. Future studies with a larger sample and additional participating sites are recommended.

12.
Chem Res Toxicol ; 30(2): 574-582, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28027439

ABSTRACT

Carbaryl (1-naphthyl methylcarbamate) and carbofuran (2,3-dihydro-2,2-dimethyl-7-benzofuranyl methylcarbamate) are among the most toxic insecticides, implicated in a variety of diseases including diabetes and cancer among others. Using an integrated pharmacoinformatics based screening approach, we have identified these insecticides to be structural mimics of the neurohormone melatonin and were able to bind to the putative melatonin binding sites in MT1 and MT2 melatonin receptors in silico. Carbaryl and carbofuran then were tested for competition with 2-[125I]-iodomelatonin (300 pM) binding to hMT1 or hMT2 receptors stably expressed in CHO cells. Carbaryl and carbofuran showed higher affinity for competition with 2-[125I]-iodomelatonin binding to the hMT2 compared to the hMT1 melatonin receptor (33 and 35-fold difference, respectively) as predicted by the molecular modeling. In the presence of GTP (100 µM), which decouples the G-protein linked receptors to modulate signaling, the apparent efficacy of carbaryl and carbofuran for 2-[125I]-iodomelatonin binding for the hMT1 melatonin receptor was not affected but significantly decreased for the hMT2 melatonin receptor compatible with receptor antagonist/inverse agonist and agonist efficacy, respectively. Altogether, our data points to a potentially new mechanism through which carbamate insecticides carbaryl and carbofuran could impact human health by altering the homeostatic balance of key regulatory processes by directly binding to melatonin receptors.


Subject(s)
Carbamates/toxicity , Insecticides/toxicity , Receptors, Melatonin/drug effects , Animals , Binding Sites , CHO Cells , Carbamates/metabolism , Cricetinae , Cricetulus , Humans , Insecticides/metabolism , Ligands , Molecular Docking Simulation , Receptors, Melatonin/metabolism
13.
FASEB J ; 31(1): 388-399, 2017 01.
Article in English | MEDLINE | ID: mdl-27733449

ABSTRACT

The suprachiasmatic nucleus (SCN)-often referred to as the master circadian clock-is essential in generating physiologic rhythms and orchestrating synchrony among circadian clocks. This study tested the hypothesis that periodic motivation induced by rhythmically pairing 2 reinforcing stimuli [methamphetamine (Meth) and running wheel (RW)] restores autonomous circadian activity in arrhythmic SCN-lesioned (SCNX) C3H/HeN mice. Sham-surgery and SCNX mice were treated with either Meth (1.2 mg/kg, i.p.) or vehicle in association, dissociation, or absence of an RW. Only the association of Meth treatment and restricted RW access successfully reestablished entrained circadian rhythms in mice with SCNX. RW-likely acting as a link between the circadian and reward systems-promotes circadian entrainment of activity. We conclude that a conditioned drug response is a powerful tool to entrain, drive, and restore circadian physiology. Furthermore, an RW should be recognized as a potent input signal in addition to the conventional use as an output signal.-Rawashdeh, O., Clough, S. J., Hudson, R. L., Dubocovich, M. L. Learned motivation drives circadian physiology in the absence of the master circadian clock.


Subject(s)
Circadian Rhythm/physiology , Learning/physiology , Motivation/physiology , Suprachiasmatic Nucleus/pathology , Animals , Central Nervous System Stimulants/pharmacology , Circadian Rhythm/drug effects , Male , Methamphetamine/pharmacology , Mice , Mice, Inbred C3H , Motivation/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Suprachiasmatic Nucleus/physiology
14.
Br J Pharmacol ; 173(18): 2702-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27314810

ABSTRACT

Melatonin receptors are seven transmembrane-spanning proteins belonging to the GPCR superfamily. In mammals, two melatonin receptor subtypes exist - MT1 and MT2 - encoded by the MTNR1A and MTNR1B genes respectively. The current review provides an update on melatonin receptors by the corresponding subcommittee of the International Union of Basic and Clinical Pharmacology. We will highlight recent developments of melatonin receptor ligands, including radioligands, and give an update on the latest phenotyping results of melatonin receptor knockout mice. The current status and perspectives of the structure of melatonin receptor will be summarized. The physiological importance of melatonin receptor dimers and biologically important and type 2 diabetes-associated genetic variants of melatonin receptors will be discussed. The role of melatonin receptors in physiology and disease will be further exemplified by their functions in the immune system and the CNS. Finally, antioxidant and free radical scavenger properties of melatonin and its relation to melatonin receptors will be critically addressed.


Subject(s)
Receptors, Melatonin , Animals , Humans , Ligands , Receptors, Melatonin/chemistry , Receptors, Melatonin/deficiency , Receptors, Melatonin/immunology
15.
Synapse ; 70(5): 181-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26799638

ABSTRACT

Melatonin (MLT) is secreted from the pineal gland and mediates its physiological effects through activation of two G protein-coupled receptors, MT1 and MT2 . These receptors are expressed in several brain areas, including the habenular complex, a pair of nuclei that relay information from forebrain to midbrain and modulate a plethora of behaviors, including sleep, mood, and pain. However, so far, the precise mechanisms by which MLT control the function of habenula neurons remain unknown. Using whole cell recordings from male rat brain slices, we examined the effects of MLT on the excitability of medial lateral habenula (MLHb) neurons. We found that MLT had no significant effects on the intrinsic excitability of MLHb neurons, but profoundly increased the amplitude of glutamate-mediated evoked excitatory post-synaptic currents (EPSC). The increase in strength of glutamate synapses onto MLHb neurons was mediated by an increase in glutamate release. The MLT-induced increase in glutamatergic synaptic transmission was blocked by the competitive MT1 /MT2 receptor antagonist luzindole (LUZ). These results unravel a potential cellular mechanism by which MLT receptor activation enhances the excitability of MLHb neurons. The MLT-mediated control of glutamatergic inputs to the MLHb may play a key role in the modulation of various behaviors controlled by the habenular complex.


Subject(s)
Excitatory Postsynaptic Potentials , Glutamic Acid/metabolism , Habenula/metabolism , Receptors, Melatonin/metabolism , Synapses/metabolism , Animals , Habenula/physiology , Male , Rats , Rats, Sprague-Dawley , Receptors, Melatonin/antagonists & inhibitors , Synapses/physiology , Tryptamines/pharmacology
16.
Annu Rev Pharmacol Toxicol ; 56: 361-83, 2016.
Article in English | MEDLINE | ID: mdl-26514204

ABSTRACT

Melatonin, or 5-methoxy-N-acetyltryptamine, is synthesized and released by the pineal gland and locally in the retina following a circadian rhythm, with low levels during the day and elevated levels at night. Melatonin activates two high-affinity G protein-coupled receptors, termed MT1 and MT2, to exert beneficial actions in sleep and circadian abnormality, mood disorders, learning and memory, neuroprotection, drug abuse, and cancer. Progress in understanding the role of melatonin receptors in the modulation of sleep and circadian rhythms has led to the discovery of a novel class of melatonin agonists for treating insomnia, circadian rhythms, mood disorders, and cancer. This review describes the pharmacological properties of a slow-release melatonin preparation (i.e., Circadin®) and synthetic ligands (i.e., agomelatine, ramelteon, tasimelteon), with emphasis on identifying specific therapeutic effects mediated through MT1 and MT2 receptor activation. Discovery of selective ligands targeting the MT1 or the MT2 melatonin receptors may promote the development of novel and more efficacious therapeutic agents.


Subject(s)
Melatonin/metabolism , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Animals , Circadian Rhythm/physiology , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/metabolism , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/metabolism
17.
Physiol Behav ; 132: 79-86, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24813704

ABSTRACT

The drug of abuse methamphetamine (METH) is known for its ability to enhance reward responses. The rewarding properties of psychostimulants have been shown to vary across time of day in mice. The goal of this study was to determine the role of the MT1 and MT2 melatonin receptors in METH-induced reward, as measured by the conditioned place preference (CPP) paradigm during the light and dark phases. C3H/HeN wild-type mice were trained for METH-induced CPP at either ZT 6-8 (ZT: Zeitgeber time; ZT 0=lights on), when endogenous melatonin levels are low, or ZT 19-21, when melatonin levels are high. These time points also correspond to the high and low points for expression of the circadian gene Period1, respectively. The locomotor response to METH (1.2mg/kg, ip) treatment was of similar magnitude at both times; however only C3H/HeN mice conditioned to METH at ZT 6-8 developed a place preference. C3H/HeN mice with a genetic deletion of either the MT1 (MT1KO) or MT2 (MT2KO) receptor tested at ZT 6-8 or ZT 19-21 did not develop a place preference for METH, though both showed a similar increase in locomotor activity following METH treatment when compared to wild-type mice. We conclude that in our mouse model METH-induced CPP is dependent on time of day and the presence of the MT1 or MT2 receptors, suggesting a role for melatonin in METH-induced reward.


Subject(s)
Central Nervous System Stimulants/pharmacology , Conditioning, Operant/drug effects , Methamphetamine/pharmacology , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , Reward , Analysis of Variance , Animals , Conditioning, Operant/physiology , Dose-Response Relationship, Drug , Locomotion/drug effects , Locomotion/genetics , Male , Melatonin/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Time Factors
18.
J Pineal Res ; 56(3): 254-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24446898

ABSTRACT

Depression is often linked to early-life adversity and circadian disturbances. Here, we assessed the long-term impact of early-life adversity, particularly preweaning mother-infant separation, on the circadian system's responsiveness to a time giver or synchronizer (Zeitgeber). Mother-reared (MR) and peer-reared (PR) rhesus monkeys were subjected to chronic jet-lag, a forced desynchrony protocol of 22 hr T-cycles [11:11 hr light:dark (LD) cycles] to destabilize the central circadian organization. MR and PR monkeys subjected to the T-cycles showed split locomotor activity rhythms with periods of ~22 hr (entrained) and ~24 hr (free-running), simultaneously. Continuous melatonin treatment in the drinking water (20 µg/mL) gradually increased the amplitude of the entrained rhythm at the expense of the free-running rhythm, reaching complete entrainment by 1 wk. Upon release into constant dim light, a rearing effect on anticipation for both the predicted light onset and food presentation was observed. In MR monkeys, melatonin did not affect the amplitude of anticipatory behavior. Interestingly, however, PR macaques showed light onset and food anticipatory activities in response to melatonin treatment. These results demonstrate for the first time a rearing-dependent effect of maternal separation in macaques, imprinting long-term plastic changes on the circadian system well into late adulthood. These effects could be counteracted by the synchronizer molecule melatonin. We conclude that the melatonergic system is targeted by early-life adversity of maternal separation and that melatonin supplementation ameliorates the negative impact of stress on the circadian system.


Subject(s)
Circadian Rhythm/physiology , Maternal Deprivation , Melatonin/pharmacology , Motor Activity/drug effects , Photoperiod , Stress, Psychological/physiopathology , Animals , Female , Jet Lag Syndrome/physiopathology , Macaca mulatta , Male , Neuronal Plasticity/drug effects
19.
Psychopharmacology (Berl) ; 231(1): 257-67, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23934259

ABSTRACT

RATIONALE: Melatonin modifies physiological and behavioral responses to psychostimulants, with the MT1 and MT2 melatonin receptors specifically implicated in facilitating methamphetamine (METH)-induced sensitization in melatonin-proficient mice. OBJECTIVE: The objective of the study is to assess differences in locomotor sensitization after a single dose of methamphetamine in low-melatonin-expressing C57BL/6 wild-type and MT1 receptor knockout (MT1KO) mice, comparing with melatonin-expressing C3H/HeN mice. METHODS: Mice received a vehicle or methamphetamine (1.2 mg/kg, i.p.) pretreatment (day 1) during the light (ZT5-9) or dark (ZT 19-21) periods in novel test arenas. Locomotor sensitization was assessed by methamphetamine challenge after an eight-day abstinence (day 9). TH protein expression was evaluated by immunofluorescence and Western blot analysis. RESULTS: Methamphetamine pretreatment induced statistically significant locomotor sensitization upon challenge after eight-day abstinence in C3H and C57 wild-type mice during the light period. The magnitude of sensitization in C57 mice was diminished in the dark period and completely abrogated in MT1KO mice. No differences were observed in tyrosine hydroxylase immunoreactivity in the mesolimbic dopamine system. Additional exposures to the test arenas after methamphetamine pretreatment (nights 2-6) enhanced sensitization. CONCLUSIONS: Deletion of the MT1 melatonin receptor abolishes sensitization induced by a single METH pretreatment. The magnitude of sensitization is also altered by time of day and contextual cues. We conclude that the MT1 melatonin receptor is emerging as a novel target of therapeutic intervention for drug abuse disorders.


Subject(s)
Central Nervous System Stimulants/pharmacology , Methamphetamine/pharmacology , Motor Activity/drug effects , Receptor, Melatonin, MT1/drug effects , Animals , Blotting, Western , Brain/enzymology , Circadian Rhythm/drug effects , Darkness , Light , Male , Melatonin/genetics , Melatonin/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Receptor, Melatonin, MT1/genetics , Species Specificity , Tyrosine 3-Monooxygenase/metabolism
20.
J Pineal Res ; 54(2): 222-31, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23190173

ABSTRACT

This study assessed the role of melatonin in modulating running wheel(RW)-induced hippocampal neurogenesis in adult C3H/HeN mice. Chronic melatonin (0.02 mg/mL, oral for 12 days) treatment did not affect cell proliferation or cell survival determined by the number of BrdU-positive cells in dentate gyrus of mice with access to fixed wheel (FW). RW activity significantly increased cell proliferation [RW (n = 8) versus FW (n = 6): dorsal, 199 ± 18 versus 125 ± 12, P < 0.01; ventral, 211 ± 15 versus 123 ± 13, P < 0.01] and newborn cell survival [RW (n = 7) versus FW (n = 8): dorsal, 45 ± 8.5 versus 15 ± 1.8, P < 0.01; ventral, 48 ± 8.1 versus 15 ± 1.4)] in the dorsal and ventral dentate gyrus. Oral melatonin treatment further potentiated RW activity-induced cell survival in both areas of the dentate gyrus [melatonin (n = 10) versus vehicle (n = 7): dorsal, 63 ± 5.4 versus 45 ± 8.5 P < 0.05; ventral, 75 ± 7.9 versus 48 ± 8.1, P < 0.01] and neurogenesis [melatonin (n = 8) versus vehicle (n = 8): dorsal, 46 ± 3.4, versus 34 ± 4.5, P < 0.05; ventral, 41 ± 3.4 versus 25 ± 2.4, P < 0.01]. We conclude that melatonin potentiates RW-induced hippocampal neurogenesis by enhancing neuronal survival suggesting that the combination of physical exercise and melatonin may be an effective treatment for diseases affecting the hippocampus neurogenesis.


Subject(s)
Dentate Gyrus/cytology , Melatonin/therapeutic use , Neuralgia/drug therapy , Neurogenesis/drug effects , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Neuralgia/metabolism , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...