Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
J Pineal Res ; 67(2): e12580, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30968433

ABSTRACT

Recent studies have highlighted the involvement of melatonin in the regulation of energy homeostasis. In this study, we report that mice lacking melatonin receptor 1 (MT1 KO) gained more weight, had a higher cumulative food intake, and were more hyperphagic after fasting compared to controls (WT). In response to a leptin injection, MT1 KO mice showed a diminished reduction in body weight and food intake. To evaluate hypothalamic leptin signaling, we tested leptin-induced phosphorylation of the signal transducer and activator of transcription 3 (STAT3). Leptin failed to induce STAT3 phosphorylation in MT1 KO mice beyond levels observed in mice injected with phosphate-buffered saline (PBS). Furthermore, STAT3 phosphorylation within the arcuate nucleus (ARH) was decreased in MT1 KO mice. Leptin receptor mRNA levels in the hypothalamus of MT1 KO were significantly reduced (about 50%) compared to WT. This study shows that: (a) MT1 deficiency causes weight gain and increased food intake; (b) a lack of MT1 signaling induces leptin resistance; (c) leptin resistance is ARH region-specific; and (d) leptin resistance is likely due to down-regulation of the leptin receptor. Our data demonstrate that MT1 signaling is an important modulator of leptin signaling.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Leptin/metabolism , Receptor, Melatonin, MT1/deficiency , Signal Transduction , Animals , Gene Deletion , Leptin/genetics , Male , Mice , Mice, Knockout , Receptor, Melatonin, MT1/metabolism
4.
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
5.
Exp Eye Res ; 165: 90-95, 2017 12.
Article in English | MEDLINE | ID: mdl-28941766

ABSTRACT

Earlier studies in Xenopus have indicated a role for melatonin in the regulation of retinal disk shedding, but the role of melatonin in the regulation of daily rhythm in mammalian disk shedding and phagocytosis is still unclear. We recently produced a series of transgenic mice lacking melatonin receptor type 1 (MT1) or type 2 (MT2) in a melatonin-proficient background and have shown that removal of MT1 and MT2 receptors induces significant effects on daily and circadian regulation of the electroretinogram as well as on the viability of photoreceptor cells during aging. In this study we investigated the daily rhythm of phagocytic activity by the retinal pigment epithelium in MT1 and MT2 knock-out mice. Our data indicate that in MT1 and MT2 knock-out mice the peak of phagocytosis is advanced by 3 h with respect to wild-type mice and occurred in dark rather than after the onset of light, albeit the mean phagocytic activity over the 24-h period did not change among the three genotypes. Nevertheless, this small change in the profile of daily phagocytic rhythms may produce a significant effect on retinal health since MT1 and MT2 knock-out mice showed a significant increase in lipofuscin accumulation in the retinal pigment epithelium.


Subject(s)
Circadian Rhythm/physiology , Melatonin/physiology , Phagocytosis/physiology , Retinal Pigment Epithelium/physiology , Signal Transduction/physiology , Animals , Disease Models, Animal , Electroretinography , Mice , Mice, Inbred C3H , Mice, Knockout , Mice, Transgenic , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency
6.
Int J Mol Sci ; 18(3)2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28257037

ABSTRACT

The role of endogenous melatonin for the control of the circadian system under entrained conditions and for the determination of the chronotype is still poorly understood. Mice with deletions in the melatoninergic system (melatonin deficiency or the lack of melatonin receptors, respectively) do not display any obvious defects in either their spontaneous (circadian) or entrained (diurnal) rhythmic behavior. However, there are effects that can be detected by analyzing the periodicity of the locomotor behaviors in some detail. We found that melatonin-deficient mice (C57Bl), as well as melatonin-proficient C3H mice that lack the melatonin receptors (MT) 1 and 2 (C3H MT1,2 KO), reproduce their diurnal locomotor rhythms with significantly less accuracy than mice with an intact melatoninergic system. However, their respective chronotypes remained unaltered. These results show that one function of the endogenous melatoninergic system might be to stabilize internal rhythms under conditions of a steady entrainment, while it has no effects on the chronotype.


Subject(s)
Behavior, Animal , Light , Melatonin/biosynthesis , Animals , Circadian Rhythm , Male , Melatonin/deficiency , Mice , Mice, Knockout , Motor Activity , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism
7.
Neuroendocrinology ; 105(1): 35-43, 2017.
Article in English | MEDLINE | ID: mdl-27490331

ABSTRACT

BACKGROUND/METHODS: Melatonin, the neurohormone for darkness, mediates photoperiod-dependent changes in physiology and behavior by targeting specific membrane-bound receptors (MT1 and MT2). In the present study, we investigated the impact of MT1 receptor deficiency on feeding behavior, locomotor activity and mRNA expression levels encoding for the polypeptide pro-opiomelanocortin (Pomc) and neuropeptide Y (Npy) in the hypothalamic arcuate nucleus (ARC) and the adenohypophysis [pars distalis (PD) and pars intermedia (PI)] in a comparison between wild-type (WT) and MT1-deficient (MT1-/-) mice. RESULTS: The MT1-/- mice spent significantly more time feeding than the WT mice, while the general locomotor behavior, body weight and the total amount of food consumed did not differ between both genotypes. The nocturnal expression levels of Pomc in the ARC and PD were significantly higher in WT as compared to MT1-/- mice and exogenous melatonin administered during the light phase stimulated Pomc expression in WT mice only. No differences were found between WT and MT1-/- mice with regard to Pomc expression levels in the PI. CONCLUSION: Thus, the MT1-mediated signaling stimulates Pomc expression in a region-specific pattern. Since the MT1-mediated changes in Pomc expression do not elicit direct orexigenic or anorexigenic effects, such effects are obviously mediated by regulatory systems downstream of the Pomc mRNA (e.g. cleavage and release of POMC derivatives), which are independent of MT1 signaling.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Feeding Behavior/physiology , Gene Expression Regulation/genetics , Pituitary Gland/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Melatonin, MT1/deficiency , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Eating/drug effects , Eating/genetics , Feeding Behavior/drug effects , Locomotion/drug effects , Locomotion/genetics , Male , Melatonin/pharmacology , Mice , Mice, Knockout , Neuropeptide Y/metabolism , Pituitary Gland/drug effects , Pro-Opiomelanocortin/genetics , Receptor, Melatonin, MT1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
8.
PLoS One ; 11(10): e0164665, 2016.
Article in English | MEDLINE | ID: mdl-27727308

ABSTRACT

The energy metabolism of the retina might comply with daily changes in energy demand and is impaired in diabetic retinopathy-one of the most common causes of blindness in Europe and the USA. The aim of this study was to investigate putative adaptation of energy metabolism in healthy and diabetic retina. Hence expression analysis of metabolic pathway genes was performed using quantitative polymerase chain reaction, semi-quantitative western blot and immunohistochemistry. Transcriptional profiling of key enzymes of energy metabolism identified transcripts of mitochondrial fatty acid ß-oxidation enzymes, i.e. carnitine palmitoyltransferase-1α (Cpt-1α) and medium chain acyl-CoA dehydrogenase (Acadm) to display daily rhythms with peak values during daytime in preparations of the whole retina and microdissected photoreceptors. The cycling of both enzymes persisted in constant darkness, was dampened in mice deficient for dopamine D4 (D4) receptors and was altered in db/db mice-a model of diabetic retinopathy. The data of the present study are consistent with circadian clock-dependent and dopaminergic regulation of fatty acid oxidation in retina and its putative disturbance in diabetic retina.


Subject(s)
Circadian Rhythm/physiology , Dopamine/metabolism , Fatty Acids/metabolism , Photoreceptor Cells/metabolism , Retina/metabolism , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Energy Metabolism , Fatty Acids/chemistry , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Microscopy, Fluorescence , Oxidation-Reduction , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Receptors, Dopamine D4/deficiency , Receptors, Dopamine D4/genetics
9.
Cell Tissue Res ; 365(1): 147-56, 2016 07.
Article in English | MEDLINE | ID: mdl-26917036

ABSTRACT

Ecto-5'-nucleotidase (eN) is the major extracellular adenosine-producing ecto-enzyme in mouse brain. Via the production of adenosine, eN participates in many physiological and pathological processes, such as wakefulness, inflammation, nociception and neuroprotection. The mechanisms regulating the expression of eN are therefore of considerable neurobiological and clinical interest. Having previously described a modulatory effect of melatonin in the regulation of eN mRNA levels, we decided to analyze the melatonin receptor subtype involved in the regulation of eN mRNA levels by comparing eN mRNA patterns in melatonin-proficient transgenic mice lacking either the melatonin receptor subtype 1 (MT1 KO) or both melatonin receptor subtypes (MT1 and MT2; MT1/2 KO) with the corresponding melatonin-proficient wild-type (WT) controls. By means of radioactive in situ hybridization, eN mRNA levels were found to be diminished in both MT1 and MT1/2 KO mice compared with WT controls suggesting stimulatory impacts of melatonin receptors on eN mRNA levels. Whereas eN mRNA levels increased during the day and peaked at night in WT and MT1 KO mice, eN mRNA levels at night were reduced and the peak was shifted toward day-time in double MT1/2 KO mice. These data suggest that the MT2 receptor subtype may play a role in the temporal regulation of eN mRNA availability. Notably, day-time locomotor activity was significantly higher in MT1/2 KO compared with WT mice. Our results suggest melatoninergic signaling as an interface between the purinergic system and the circadian system.


Subject(s)
5'-Nucleotidase/genetics , Prosencephalon/metabolism , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , 5'-Nucleotidase/metabolism , Animals , Mice , Mice, Knockout , Motor Activity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Time Factors
10.
PLoS One ; 11(1): e0148214, 2016.
Article in English | MEDLINE | ID: mdl-26824606

ABSTRACT

Melatonin is rhythmically secreted by both the pineal gland and retina in a circadian fashion, with its peak synthesis occurring during the night. Once synthesized, melatonin exerts its effects by binding to two specific G-protein coupled receptors-melatonin receptor type 1(MT1) and melatonin receptor type 2(MT2). Recent studies suggest the involvement of MT1 and MT2 in the regulation of glucose homeostasis; however the ability of melatonin signaling to impart timing cues on glucose metabolism remains poorly understood. Here we report that the removal of MT1 or MT2 in mice abolishes the daily rhythm in blood glucose levels. Interestingly, removal of melatonin receptors produced small effects on the rhythmic expression patterns of clock genes within skeletal muscle, liver, and adipose tissue. Taken together, our data suggest that the loss of the daily rhythm in blood glucose observed in MT1(-/-) and MT2(-/-) mice does not occur as a consequence of 'disrupted' clocks within insulin sensitive tissues. Finally our results highlight a diurnal contribution of melatonin receptor signaling in the daily regulation of blood glucose levels.


Subject(s)
Blood Glucose/metabolism , Circadian Rhythm/genetics , Melatonin/metabolism , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Adipose Tissue/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Gene Expression Regulation , Homeostasis , Liver/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Pineal Gland/metabolism , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , Retina/metabolism , Signal Transduction
11.
Int J Neuropsychopharmacol ; 18(3)2015 Jan 31.
Article in English | MEDLINE | ID: mdl-25638817

ABSTRACT

BACKGROUND: Melancholic depression, described also as endogenous depression, is a mood disorder with distinctive specific psychopathological features and biological homogeneity, including anhedonia, circadian variation of mood, psychomotor activation, weight loss, diurnal cortisol changes, and sleep disturbances. Although several hypotheses have been proposed, the etiology of this disorder is still unknown. METHODS: Behavioral, electrophysiological and biochemical approaches were used to characterize the emotional phenotype, serotonergic and noradrenergic electrical activity, and corticosterone in melatonin MT1 receptor knockout mice and their wild type counterparts, during both light and dark phases. RESULTS: Melatonin MT1 receptor knockout mice have decreased mobility in the forced swim and tail suspension tests as well as decreased sucrose consumption, mostly during the dark/inactive phase. These mood variations are reversed by chronic treatment with the tricyclic antidepressant desipramine. In addition, MT1 receptor knockout mice exhibit psychomotor disturbances, higher serum levels of corticosterone the dark phase, and a blunted circadian variation of corticosterone levels. In vivo electrophysiological recordings show a decreased burst-firing activity of locus coeruleus norepinephrine neurons during the dark phase. The circadian physiological variation in the spontaneous firing activity of high-firing neuronal subpopulations of both norepinephrine neurons and dorsal raphe serotonin neurons are abolished in MT1 knockout mice. CONCLUSIONS: These data demonstrate that melatonin MT1 receptor knockout mice recapitulate several behavioral and neurobiological circadian changes of human melancholic depression and, for the first time, suggest that the MT1 receptor may be implicated in the pathogenesis of melancholic depression and is a potential pharmacological target for this mental condition.


Subject(s)
Chronobiology Disorders/genetics , Circadian Rhythm/genetics , Depressive Disorder/genetics , Receptor, Melatonin, MT1/deficiency , Animals , Antidepressive Agents, Tricyclic/therapeutic use , Chronobiology Disorders/drug therapy , Corticosterone/blood , Depressive Disorder/drug therapy , Desipramine/therapeutic use , Disease Models, Animal , Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Food Preferences , Hindlimb Suspension , Male , Maze Learning/drug effects , Mice , Mice, Knockout , Receptor, Melatonin, MT1/genetics , Swimming
12.
Neuroscience ; 277: 506-21, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25046530

ABSTRACT

Melatonin, an indoleamine hormone secreted into circulation at night primarily by the brain's pineal gland, has been shown to have a wide variety of actions on the development and physiology of neurons in the CNS. Acting via two G-protein-coupled membrane receptors (MT1 and MT2), melatonin modulates neurogenesis, synaptic functions, neuronal cytoskeleton and gene expression. In the present studies, we sought to characterize the behavior and neuronal biology of transgenic mice lacking both of these melatonin receptors as a way to understand the hormone's receptor versus non-receptor-mediated actions in CNS-dependent activities, such as learning and memory, anxiety, general motor performance and circadian rhythmicity. Assessment of these behaviors was complemented by molecular analyses of gene expression in the brain. Our results demonstrate mild behavioral hyperactivity and a lengthened circadian period of free-running motor activity in melatonin receptor-deficient mice as compared to receptor-intact control mice beginning at an early age. Significant improvement in cognitive performance was found using the Barnes Maze and the Y-Maze. No behavioral changes in anxiety levels were found. Electrophysiological measures in hippocampal slices revealed a clear enhancement of long-term potentiation in mice lacking melatonin receptors with no significant differences in paired-pulse facilitation. Quantitative analysis of brain protein expression levels of phosphoCREB and phosphoERK1/2 and key markers of synaptic activity (synapsin, glutamate receptor 1, spinophilin, and glutamic acid decarboxylase 1) revealed significant differences between the double-knockout and wild-type animals, consistent with the behavioral findings. Thus, genetic deletion of melatonin receptors produces mice with enhanced cognitive and motor performance, supporting the view that these receptors play an important role in neurobehavioral development.


Subject(s)
Frontal Lobe/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , Animals , Anxiety/physiopathology , CREB-Binding Protein/metabolism , Circadian Rhythm/physiology , Drinking/physiology , Locomotion/physiology , MAP Kinase Signaling System/physiology , Male , Melatonin/blood , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Random Allocation , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics
13.
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
14.
Curr Aging Sci ; 6(1): 125-33, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23895529

ABSTRACT

The pineal hormone melatonin (MLT) has potent anti-breast cancer activity, its actions are heavily mediated via the MT1 receptor and subsequent modulation of downstream signaling pathways including cAMP/PKA, Erk/MAPK, p38, and Ca2+/calmodulin. Also, via the MT1 pathway, MLT can repress the transcriptional activity of some mitogenic nuclear receptors including ERα, GR, and RORα, while potentiating the activity of other receptors (RARα and RXRα) involved in differentiation, anti-proliferation, and apoptosis. A review of the literature supports the view that MLT, via its MT1 receptor, can suppress all phases of breast cancer including initiation, promotion, and progression. During the fifth and sixth decades of life, the production of MLT diminishes, concurrently with an increase in the incidence of breast cancer. Inasmuch as MLT has been demonstrated to have anti-cancer activity, we hypothesized that there may be a causal link between the reduction in MLT production in the pineal gland and the incidence of breast cancer which increases with age. We designed this study to establish whether a truly inverse relationship exists between tissue-isolated mammary tumor growth in young (2 months), adult (12 months), and old (20 months) female Buffalo rats and the decrease in both MLT and the MT1 receptor with age, such that a causal link could be found. Serum MLT levels were measured in both the light and dark phases. A significant 29% decrease in serum MLT levels, measured at the nocturnal peak, was found in the adult and senescent rats (75% decrease) in comparison to that in young rats. In young rats, the nocturnal pineal gland MLT content exceeded daytime levels by 19-fold compared to a sevenfold increase in old mice. Also, the MT1 receptor was found to be significantly lower in the nighttime and early morning in the senescent rat uterus as compared to uteri from young and adult rats. Analysis of the rate of growth in transplanted, tissue-isolated, mammary tumors induced by N-nitroso-n-methyl-urea (NMU) showed a significant increase in the senescent rats, but not in the young or adult rats Additionally, diminished response to the inhibitory action on tumor growth of exogenous MLT was noted in senescent rats such that tumor growth was suppressed by only 33% compared to 48% and 66% in adult and young rats, respectively. The diminution of the response of tumors to exogenous MLT was found to correlate with reduced MT1 receptor expression in senescent compared to young and adult rats. These data suggest that the observed age-associated enhanced growth of tumors is related to the much reduced levels of MLT and its receptor in aged animals which reduce the sensitivity of tumors to inhibition by exogenous MLT.


Subject(s)
Aging/physiology , Mammary Neoplasms, Experimental/etiology , Melatonin/deficiency , Receptor, Melatonin, MT1/deficiency , Animals , Breast Neoplasms/etiology , Breast Neoplasms/physiopathology , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/physiopathology , Melatonin/physiology , Melatonin/therapeutic use , Mice , Pineal Gland/physiopathology , Rats , Receptor, Melatonin, MT1/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Uterus/physiopathology
15.
Behav Brain Res ; 243: 231-8, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23333399

ABSTRACT

The neurohormone melatonin activates two G-protein coupled receptors, MT1 and MT2. Melatonin is implicated in circadian rhythms and sleep regulation, but the role of its receptors remains to be defined. We have therefore characterized the spontaneous vigilance states in wild-type (WT) mice and in three different types of transgenic mice: mice with genetic inactivation of MT1 (MT1(-/-)), MT2 (MT2(-/-)) and both MT1/MT2 (MT1(-/-)/MT2(-/-)) receptors. Electroencephalographic (EEG) and electromyographic sleep-wake patterns were recorded across the 24-h light-dark cycle. MT1(-/-)mice displayed a decrease (-37.3%) of the 24-h rapid eye movement sleep (REMS) time whereas MT2(-/-)mice showed a decrease (-17.3%) of the 24-h non rapid eye movement sleep (NREMS) time and an increase in wakefulness time (14.8%). These differences were the result of changes occurring in particular during the light/inactive phase. Surprisingly, MT1(-/-)/MT2(-/-) mice showed only an increase (8.9%) of the time spent awake during the 24-h. These changes were correlated to a decrease of the REMS EEG theta power in MT1(-/-)mice, of the NREMS EEG delta power in MT2(-/-)mice, and an increase of the REMS and wakefulness EEG theta power in MT1(-/-)/MT2(-/-) mice. Our results show that the genetic inactivation of both MT1 and MT2 receptors produces an increase of wakefulness, likely as a result of reduced NREMS due to the lack of MT2 receptors, and reduced REMS induced by the lack of MT1 receptors. Therefore, each melatonin receptor subtype differently regulates the vigilance states: MT2 receptors mainly NREMS, whereas MT1 receptors REMS.


Subject(s)
Cerebral Cortex/physiology , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/physiology , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/physiology , Sleep Stages/genetics , Wakefulness/genetics , Animals , Cerebral Cortex/pathology , Delta Rhythm/genetics , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Mice, Transgenic , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Sleep, REM/genetics , Theta Rhythm/genetics
16.
J Pineal Res ; 53(4): 390-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22672634

ABSTRACT

Melatonin has been shown to modulate glucose metabolism by influencing insulin secretion. Recent investigations have also indicated a regulatory function of melatonin on the pancreatic α-cells. The present in vitro and in vivo studies evaluated whether melatonin mediates its effects via melatonin receptors and which signaling cascade is involved. Incubation experiments using the glucagon-producing mouse pancreatic α-cell line αTC1 clone 9 (αTC1.9) as well as isolated pancreatic islets of rats and mice revealed that melatonin increases glucagon secretion. Preincubation of αTC1.9 cells with the melatonin receptor antagonists luzindole and 4P-PDOT abolished the glucagon-stimulatory effect of melatonin. In addition, glucagon secretion was lower in the pancreatic islets of melatonin receptor knockout mice than in the islets of the wild-type (WT) control animals. Investigations of melatonin receptor knockout mice revealed decreased plasma glucagon concentrations and elevated mRNA expression levels of the hepatic glucagon receptor when compared to WT mice. Furthermore, studies using pertussis toxin, as well as measurements of cAMP concentrations, ruled out the involvement of Gαi- and Gαs-coupled signaling cascades in mediating the glucagon increase induced by melatonin. In contrast, inhibition of phospholipase C in αTC1.9 cells prevented the melatonin-induced effect, indicating the physiological relevance of the Gαq-coupled pathway. Our data point to the involvement of the phosphatidylinositol 3-kinase signaling cascade in mediating melatonin effects in pancreatic α-cells. In conclusion, these findings provide evidence that the glucagon-stimulatory effect of melatonin in pancreatic α-cells is melatonin receptor mediated, thus supporting the concept of melatonin-modulated and diurnal glucagon release.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon/metabolism , Melanins/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Receptor, Melatonin, MT1/drug effects , Receptor, Melatonin, MT2/drug effects , Signal Transduction/drug effects , Animals , Cell Line , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/enzymology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Glucagon/blood , Glucagon-Secreting Cells/enzymology , Glucagon-Secreting Cells/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Pertussis Toxin/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/genetics , Receptors, Glucagon/drug effects , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Tetrahydronaphthalenes/pharmacology , Tissue Culture Techniques , Tryptamines/pharmacology , Type C Phospholipases/metabolism
17.
J Pineal Res ; 53(4): 399-409, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22672659

ABSTRACT

This study explored the role of the melatonin receptors in methamphetamine (METH)-induced locomotor sensitization during the light and dark phases in C3H/HeN mice with genetic deletion of the MT(1) and/or MT(2) melatonin receptors. Six daily treatments with METH (1.2 mg/kg, i.p.) in a novel environment during the light phase led to the development of locomotor sensitization in wild-type (WT), MT(1)KO and MT(2)KO mice. Following four full days of abstinence, METH challenge (1.2 mg/kg, i.p.) triggered the expression of locomotor sensitization in METH-pretreated but not in vehicle (VEH)-pretreated mice. In MT(1)/MT(2)KO mice, the development of sensitization during the light phase was significantly reduced and the expression of sensitization was completely abrogated upon METH challenge. During the dark phase the development of locomotor sensitization in METH-pretreated WT, MT(1)KO and MT(2)KO mice was statistically different from VEH-treated controls. However, WT and MT(2)KO, but not MT(1)KO mice receiving repeated VEH pretreatments during the dark phase expressed a sensitized response to METH challenge that is of an identical magnitude to that observed upon 6 days of METH pretreatment. We conclude that exposure to a novel environment during the dark phase, but not during the light phase, facilitated the expression of sensitization to a METH challenge in a manner dependent on MT(1) melatonin receptor activation by endogenous melatonin. We suggest that MT(1) and MT(2) melatonin receptors are potential targets for pharmacotherapeutic intervention in METH abusers.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Sensitization/drug effects , Central Nervous System Stimulants/pharmacology , Central Nervous System/drug effects , Gene Deletion , Methamphetamine/pharmacology , Motor Activity/drug effects , Photoperiod , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT2/deficiency , Animals , Behavior, Animal/radiation effects , Central Nervous System/metabolism , Central Nervous System/radiation effects , Central Nervous System Sensitization/radiation effects , Male , Melatonin/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Motor Activity/radiation effects , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Time Factors
18.
Chronobiol Int ; 29(4): 415-29, 2012 May.
Article in English | MEDLINE | ID: mdl-22489607

ABSTRACT

The indolamine melatonin is an important rhythmic endocrine signal in the circadian system. Exogenous melatonin can entrain circadian rhythms in physiology and behavior, but the role of endogenous melatonin and the two membrane-bound melatonin receptor types, MT1 and MT2, in reentrainment of daily rhythms to light-induced phase shifts is not understood. The present study analyzed locomotor activity rhythms and clock protein levels in the suprachiasmatic nuclei (SCN) of melatonin-deficient (C57BL/6J) and melatonin-proficient (C3H/HeN) mice, as well as in melatonin-proficient (C3H/HeN) mice with targeted deletion of the MT1, MT2, or both receptors, to determine effects associated with phase delays or phase advances of the light/dark (LD) cycle. In all mouse strains and genotypes, reentrainment of locomotor activity rhythms was significantly faster after a 6-h phase delay than a 6-h phase advance. Reentrainment after the phase advance was, however, significantly slower than in melatonin-deficient animals and in mice lacking functional MT2 receptors than melatonin-proficient animals with intact MT2 receptors. To investigate whether these behavioral differences coincide with differences in reentrainment of clock protein levels in the SCN, mPER1, mCRY1 immunoreactions were compared between control mice kept under the original LD cycle and killed at zeitgeber time 04 (ZT04) or at ZT10, respectively, and experimental mice subjected to a 6-h phase advance of the LD cycle and sacrificed at ZT10 on the third day after phase advance. This ZT corresponds to ZT04 of the original LD cycle. Under the original LD cycle, the numbers of mPER1- and mCRY1-immunoreactive cell nuclei were low at ZT04 and high at ZT10 in the SCN of all mouse strains and genotypes investigated. Notably, mouse strains with intact melatonin signaling and functional MT2 receptors showed a significant increase in the number of mPER1- and mCRY1-immunoreactive cell nuclei at the new ZT10 as compared to the former ZT04. These data suggest the endogenous melatonin signal facilitates reentrainment of the circadian system to phase advances on the level of the SCN molecular clockwork by acting upon MT2 receptors.


Subject(s)
Behavior, Animal , Circadian Rhythm , Melatonin/metabolism , Motor Activity , Photoperiod , Receptor, Melatonin, MT2/metabolism , Signal Transduction , Suprachiasmatic Nucleus/metabolism , Animals , Biological Clocks , Cryptochromes/metabolism , Disease Models, Animal , Immunohistochemistry , Jet Lag Syndrome/metabolism , Jet Lag Syndrome/physiopathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/metabolism , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/deficiency , Receptor, Melatonin, MT2/genetics , Suprachiasmatic Nucleus/physiopathology , Time Factors
19.
Gen Comp Endocrinol ; 177(1): 98-103, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22426341

ABSTRACT

The hypophysial pars tuberalis (PT) is an important interface between neuroendocrine brain centers (hypothalamus, pineal organ) and the anterior lobe of the hypophysis (PD). The best investigated role of the PT is the control of seasonally changing functions. In mammals, melatonin secreted from the pineal organ represents a major input signal to the PT. By acting upon melatonin type 1 receptors (MT1) melatonin controls the functional activity of the PT. Most interestingly, the PT sends its output signals in two directions: via a "retrograde" pathway to the hypothalamus and via an "anterograde" pathway to the PD. TSH has been identified as "retrograde" messenger, while endocannabinoids function as messengers of the "anterograde" pathway. Here we show in mice that the PT expresses Tafa-3 encoding for a secretory peptide. In the PT of wild type mice Tafa-3 mRNA levels varied between day and night: they were low at mid-day and high at mid-night. This day/night difference was not observed in the PT of mice with a targeted deletion of the MT1 receptor indicating that Tafa-3 mRNA expression in the PT is controlled by melatonin acting through the MT1 receptor. Notably, Tafa-3 expression was not restricted to the PT, but was also found in other brain regions, such as the hippocampus, the habenular and thalamic nuclei. In these regions, Tafa-3 expression did not display a day/night difference and was not affected by MT1-deficiency. Thus, Tafa-3 expression appears to be controlled by region-specific mechanisms. Our data suggest that TAFA-3 is a signaling molecule from the PT and provides further evidence for the emerging concept that the PT rather than relying upon highly organ-specific messengers employs a cocktail of signaling molecules that also operate in other brain systems.


Subject(s)
Neuropeptides/metabolism , Pituitary Gland/metabolism , Receptor, Melatonin, MT1/deficiency , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , In Situ Hybridization , Mice , Mice, Mutant Strains , Neuropeptides/genetics , Real-Time Polymerase Chain Reaction , Receptor, Melatonin, MT1/genetics
20.
Proc Natl Acad Sci U S A ; 106(35): 15043-8, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706469

ABSTRACT

A clear demonstration of the role of melatonin and its receptors in specific retinal functions is lacking. The present study investigated the distribution of MT1 receptors within the retina, and the scotopic and photopic electroretinograms (ERG) and retinal morphology in wild-type (WT) and MT1 receptor-deficient mice. MT1 receptor transcripts were localized in photoreceptor cells and in some inner retinal neurons. A diurnal rhythm in the dark-adapted ERG responses was observed in WT mice, with higher a- and b-wave amplitudes at night, but this rhythm was absent in mice lacking MT1 receptors. Injection of melatonin during the day decreased the scotopic response threshold and the amplitude of the a- and b-waves in the WT mice, but not in the MT1(-/-) mice. The effects of MT1 receptor deficiency on retinal morphology was investigated at three different ages (3, 12, and 18 months). No differences between MT1(-/-) and WT mice were observed at 3 months of age, whereas at 12 months MT1(-/-) mice have a significant reduction in the number of photoreceptor nuclei in the outer nuclear layer compared with WT controls. No differences were observed in the number of cells in inner nuclear layer or in ganglion cells at 12 months of age. At 18 months, the loss of photoreceptor nuclei in the outer nuclear layer was further accentuated and the number of ganglion cells was also significantly lower than that of controls. These data demonstrate the functional significance of melatonin and MT1 receptors in the mammalian retina and create the basis for future studies on the therapeutic use of melatonin in retinal degeneration.


Subject(s)
Melatonin/metabolism , Receptor, Melatonin, MT1/metabolism , Retina/cytology , Retina/metabolism , Vision, Ocular , Adaptation, Biological , Aging , Animals , Cell Survival , Darkness , Electroretinography , Gene Expression Regulation , Mice , Mice, Knockout , RNA, Messenger/genetics , Receptor, Melatonin, MT1/deficiency , Receptor, Melatonin, MT1/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...