Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell Mol Neurobiol ; 41(7): 1589-1598, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32734322

ABSTRACT

Melatonin is released by the pineal gland and can modulate cardiovascular system function via the G protein-coupled melatonin receptors MT1 and MT2. Most vessels are surrounded by perivascular adipose tissue (PVAT), which affects their contractility. The aim of our study was to evaluate mRNA and protein expression of MT1 and MT2 in the mesenteric artery (MA) and associated PVAT of male rats by RT-PCR and Western blot. Receptor localization was further studied by immunofluorescence microscopy. Effects of melatonin on neurogenic contractions were explored in isolated superior MA ex vivo by measurement of isometric contractile tension. MT1, but not MT2, was present in MA, and MT1 was localized mainly in vascular smooth muscle. Moreover, we proved the presence of MT1, but not MT2 receptors, in MA-associated PVAT. In isolated superior MA with intact PVAT, neuro-adrenergic contractile responses were significantly smaller when compared to arteries with removed PVAT. Pre-treatment with melatonin of PVAT-stripped arterial rings enhanced neurogenic contractions, while the potentiating effect of melatonin was not detected in preparations with preserved PVAT. We hypothesize that melatonin can stimulate the release of PVAT-derived relaxing factor(s) via MT1, which can override the direct pro-contractile effect of melatonin on vascular smooth muscle. Our results suggest that melatonin is involved in the control of vascular tone in a complex way, which is vessel specific and can reflect a sum of action on different layers of the vessel wall and surrounding PVAT.


Subject(s)
Melatonin/pharmacology , Mesenteric Arteries/metabolism , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Adipose Tissue/drug effects , Adipose Tissue/physiology , Animals , Melatonin/metabolism , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Rats, Wistar
2.
Eur J Pharmacol ; 885: 173506, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32858050

ABSTRACT

Melatonin, an emphatic endogenous molecule exerts protective effects either via activation of G-protein coupled receptors (Melatonin receptors, MTR 1-3), tumor necrosis factor receptor (TNFR), toll like receptors (TLRS), nuclear receptors (NRS) or by directly scavenging the free radicals. MTRs are extensively expressed in the heart as well as in the coronary vasculature. Accumulating evidences have indicated the existence of a strong correlation between reduction in the circulating level of melatonin and precipitation of heart attack. Apparently, melatonin exhibits cardioprotective effects via modulating inextricably interlinked pathways including modulation of mitochondrial metabolism, mitochondrial permeability transition pore formation, nitric oxide release, autophagy, generation of inflammatory cytokines, regulation of calcium transporters, reactive oxygen species, glycosaminoglycans, collagen accumulation, and regulation of apoptosis. Convincingly, this review shall describe the various signaling pathways involved in salvaging the heart against ischemia-reperfusion injury.


Subject(s)
Antioxidants/pharmacology , Melatonin/pharmacology , Myocardial Reperfusion Injury/prevention & control , Receptors, Melatonin/drug effects , Animals , Free Radical Scavengers/pharmacology , Humans , Myocardial Reperfusion Injury/pathology , Signal Transduction/drug effects
3.
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
4.
Biochem Biophys Res Commun ; 474(3): 566-571, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27130826

ABSTRACT

Melatonin, a lipophilic molecule mainly synthesized in the pineal gland, has properties of antioxidation, anti-inflammation, and antiapoptosis to improve neuroprotective functions. Here, we investigate effects and mechanisms of melatonin on neural differentiation of induced pluripotent stem cells (iPSCs). iPSCs were induced into neural stem cells (NSCs), then further differentiated into neurons in medium with or without melatonin, melatonin receptor antagonist (Luzindole) or Phosphatidylinositide 3 kinase (PI3K) inhibitor (LY294002). Melatonin significantly promoted the number of neurospheres and cell viability. In addition, Melatonin markedly up-regulated gene and protein expression of Nestin and MAP2. However, Luzindole or LY294002 attenuated these increase. The expression of pAKT/AKT were increased by Melatonin, while Luzindole or LY294002 declined these melatonin-induced increase. These results suggest that melatonin significantly increased neural differentiation of iPSCs via activating PI3K/AKT signaling pathway through melatonin receptor.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/metabolism , Melatonin/pharmacology , Neural Stem Cells/metabolism , Neurons/metabolism , Animals , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cellular Reprogramming Techniques/methods , Chromones/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice , Morpholines/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/cytology , Neurons/drug effects , Receptors, Melatonin/drug effects , Tryptamines/pharmacology
5.
Bull Exp Biol Med ; 159(1): 44-7, 2015 May.
Article in English | MEDLINE | ID: mdl-26033588

ABSTRACT

Cell damage depending on activity of quinone reductase 2 (MT3 receptor) was simulated in experiments on bone marrow cell suspension and assessed by menadione-induced DNA breaks measured by comet assay. We analyzed the protective effect of afobazole interacting with MT1, MT3, σ1 receptors, and monoamine oxidase A and its main metabolite M11 that specifi cally binds to MT3 receptors. Both compounds reduced the level of menadione-induced DNA damage (afobazole was effective in lower concentrations in comparison with M-11). Conclusion was made on the contribution of MT3 receptors to the protective effect of afobazole, but the observed concentration differences indicate possible contribution of other targets of anxiolytic drug to the protective mechanisms.


Subject(s)
Anti-Anxiety Agents/pharmacology , Benzimidazoles/pharmacology , Bone Marrow Cells/drug effects , DNA Breaks/drug effects , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Quinone Reductases/antagonists & inhibitors , Receptors, Melatonin/drug effects , Animals , Anti-Anxiety Agents/metabolism , Benzimidazoles/metabolism , Biotransformation , Cells, Cultured , Comet Assay , Dicumarol/pharmacology , Drug Evaluation, Preclinical , Metallothionein 3 , Mice , Monoamine Oxidase , Monoamine Oxidase Inhibitors , Morpholines/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Neuroprotective Agents/metabolism , Quinone Reductases/metabolism , Receptor, Melatonin, MT1/drug effects , Receptors, sigma/drug effects , Vitamin K 3/toxicity
6.
Psychopharmacology (Berl) ; 232(14): 2519-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25704105

ABSTRACT

RATIONALE: While it is known that tobacco use varies across the 24-h day, the time-of-day effects are poorly understood. Findings from several previous studies indicate a potential role for melatonin in these time-of-day effects; however, the specific underlying mechanisms have not been well characterized. Understanding of these mechanisms may lead to potential novel smoking cessation treatments. OBJECTIVE: The objective of this study is examine the role of melatonin and melatonin receptors in nicotine free-choice consumption METHODS: A two-bottle oral nicotine choice paradigm was utilized with melatonin supplementation in melatonin-deficient mice (C57BL/6J) or without melatonin supplementation in mice proficient at melatonin synthesis (C3H/Ibg) compared to melatonin-proficient mice lacking both or one of the high-affinity melatonin receptors (MT1 and MT2; double-null mutant DM, or MT1 or MT2). Preference for bitter and sweet tastants also was assessed in wild-type and MT1 and MT2 DM mice. Finally, home cage locomotor monitoring was performed to determine the effect of melatonin administration on activity patterns. RESULTS: Supplemental melatonin in drinking water significantly reduced free-choice nicotine consumption in C57BL/6J mice, which do not produce endogenous melatonin, while not altering activity patterns. Independently, genetic deletion of both MT1 and MT2 receptors in a melatonin-proficient mouse strain (C3H) resulted in significantly more nicotine consumption than controls. However, single genetic deletion of either the MT1 or MT2 receptor alone did not result in increased nicotine consumption. Deletion of MT1 and MT2 did not impact taste preference. CONCLUSIONS: This study demonstrates that nicotine consumption can be affected by exogenous or endogenous melatonin and requires at least one of the high-affinity melatonin receptors. The fact that expression of either the MT1 or MT2 melatonin receptor is sufficient to maintain lower nicotine consumption suggests functional overlap and potential mechanistic explanations.


Subject(s)
Melatonin/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Melatonin/drug effects , Animals , Choice Behavior/drug effects , Circadian Rhythm/drug effects , Drinking , Male , Melatonin/deficiency , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Receptor, Melatonin, MT1/genetics , Receptors, Melatonin/genetics , Taste/drug effects
7.
Life Sci ; 121: 158-65, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25498899

ABSTRACT

AIMS: Previous data showed that melatonin maintains liver mitochondrial homeostasis during sepsis, but neither the mechanisms underlying mitochondrial dysfunction nor the target of melatonin are known. MAIN METHODS: Here, we analyzed mitochondrial respiration in isolated mouse liver mitochondria with different substrate combinations (glutamate/malate, glutamate/malate/sucinate or succinate/rotenone) to identify mitochondrial defects and melatonin targets during sepsis. Other bioenergetic parameters including a + a3, b, and c + c1 content, mitochondrial mass, and mitochondrial supercomplexes formation were analyzed. Mitochondrial function was assessed during experimental sepsis induced by cecal ligation and puncture (CLP) in livers of 3 mo. C57BL/6 mice at early and late phases of sepsis, i.e., at 8 and 24 h after sepsis induction. KEY FINDINGS: Septic mice showed mitochondrial injury with a decrease in state 3, respiratory control rate, mitochondrial mass, and cytochrome b and c + c1 content, which was prevented by melatonin treatment. Mitochondrial dysfunction in sepsis was mainly linked to complex I damage, because complex II was far less impaired. These mitochondria preserved the respiratory supramolecular organization, maintaining their electron transport system capacity. SIGNIFICANCE: This work strengthens the use of substrate combinations to identify specific respiratory defects and selective melatonin actions in septic mitochondria. Targeting mitochondrial complex I should be a main therapeutical approach in the treatment of sepsis, whereas the use of melatonin should be considered in the therapy of clinical sepsis.


Subject(s)
Liver/metabolism , Mitochondrial Diseases/diagnosis , Receptors, Melatonin/drug effects , Sepsis/diagnosis , Animals , Citrate (si)-Synthase/metabolism , Cytochromes/metabolism , Liver/physiopathology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Diseases/physiopathology , Mitochondrial Membranes/drug effects , Oxygen Consumption/drug effects , Sepsis/metabolism , Sepsis/physiopathology , Spirometry
8.
Minerva Med ; 106(3): 169-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25000217

ABSTRACT

Delivery of melatonin and targeting melatonin receptors pose as neuroprotective strategies for stroke therapy. The potential of melatonin-based therapeutics for clinical application in stroke patients requires translational research to guide the conduct of clinical trials. We review recent preclinical and clinical data that support the use of melatonin for stroke.


Subject(s)
Melatonin/therapeutic use , Stroke/drug therapy , Clinical Trials as Topic , Humans , Receptors, Melatonin/drug effects
9.
J Neuropsychiatry Clin Neurosci ; 24(3): 290-308, 2012.
Article in English | MEDLINE | ID: mdl-23037643

ABSTRACT

Disruptions in sleep and sleep-wake cycle regulation have been identified as one of the main causes for the pathophysiology of depressive disorders. The search has been on for the identification of an ideal antidepressant that could improve both sleep disturbances and depressive symptomatology. Melatonin, the major hormone of the pineal gland, has been shown to improve sleep and is involved in the regulation of the sleep-wake cycle. Identification of high concentrations of MT1 and MT2 melatonergic receptors in the suprachiasmatic nucleus of the anterior hypothalamus, the structure concerned with regulation of circadian rhythms and sleep-wake cycles, has led to the development of melatonergic agonists with greater potency and longer durations of action. Agomelatine is one such melatonergic agonist that acts specifically on MT1/MT2 melatonergic receptors and at the same time exhibits 5-HT2C antagonism, a property that is utilized by current antidepressants that are in clinical use. Agomelatine has been shown to be effective in a number of animal models of depression. Clinical studies undertaken on patients with major depression, bipolar disorders, seasonal affective disorder, and generalized anxiety disorder have all shown that agomelatine is also very effective in ameliorating depressive symptoms and manifesting early onset of action with a good tolerability and safety profile. It improved sleep efficiency and also resynchronized the disrupted circadian rhythms. Hence, the melatonergic modulation by agomelatine is suggested as one of the mechanisms for its antidepressant effect. Agomelatine's action on dendritic neurogenesis in animal models of depression is also identified as yet another action.


Subject(s)
Acetamides/therapeutic use , Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Acetamides/chemistry , Animals , Antidepressive Agents/chemistry , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Depressive Disorder/metabolism , Depressive Disorder/pathology , Disease Models, Animal , Humans , Melatonin/metabolism , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/metabolism
10.
Reprod Fertil Dev ; 24(6): 794-803, 2012.
Article in English | MEDLINE | ID: mdl-22781930

ABSTRACT

This study aimed to investigate the effects of two different doses (100nM (M1) and 1µM (M2)) of exogenous melatonin on the reproductive capacity of Fundulus heteroclitus. Eight days of melatonin exposure significantly increased the fecundity and embryo survival of F. heteroclitus only in the M2 group compared with the control; the hatching rate was unaffected. Moreover, increases in the local expression of the melatonin receptor (mtnr) gene during follicle maturation were found; however, there were no differences between the experimental groups. Furthermore, in vitro melatonin-treated follicles showed a significantly higher germinal vesicle break down percentage compared with the control, while SDS-PAGE showed no difference in the electrophoretic pattern of the major yolk proteins. Nevertheless, densitometry revealed a greater intensity of the 118-, 95- and 40-kDa components in groups treated with melatonin. Finally, Fourier transform infrared microspectroscopy was applied to classify the different stages of oocyte development (Stages I-II, III and IV) on the basis of their macromolecular composition. The effects induced by melatonin on oogenesis were investigated by comparing vibrational spectra of females exposed to melatonin with those of controls. Changes to the Amide I band, corresponding to an increase in ß-structure, were found in oocytes of females exposed to the highest melatonin dose. These results highlight the positive role of melatonin, which is able to enhance the reproductive capacity of F. heteroclitus. Further studies are in progress to better explain the molecular mechanisms by which melatonin treatment affects reproduction in this marine species.


Subject(s)
Fundulidae/physiology , Melatonin/pharmacology , Reproduction/drug effects , Animals , Dose-Response Relationship, Drug , Egg Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Female , Fertility/drug effects , Fish Proteins/drug effects , Fish Proteins/genetics , Fish Proteins/metabolism , Fundulidae/genetics , Fundulidae/metabolism , Male , Microspectrophotometry , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Melatonin/drug effects , Receptors, Melatonin/genetics , Receptors, Melatonin/metabolism , Reproduction/genetics , Spectroscopy, Fourier Transform Infrared , Time Factors , Tissue Culture Techniques
11.
J Pineal Res ; 53(4): 335-43, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22536785

ABSTRACT

Melatonin is an indolamine that is synthesized in the pineal gland and shows a wide range of physiological functions. Although the anti-aging properties of melatonin have been reported in a senescence-accelerated mouse model, whether melatonin modulates cellular senescence has not been determined. In this study, we examined the effect of melatonin on anticancer drug-induced cellular premature senescence. We found that the doxorubicin (DOX)-induced senescence of A549 human lung cancer cells and IMR90 normal lung cells was substantially inhibited by cotreatment with melatonin in a dose-dependent manner. Mechanistically, the DOX-induced G2/M phase cell cycle arrest and the decrease in cyclinB and cdc2 expression were not affected by melatonin. However, the DOX-induced increase in intracellular levels of ROS, which is necessary for premature senescence, was completely abolished upon melatonin cotreatment. In addition, the reduction in mitochondrial membrane potential that occurs upon DOX treatment was inhibited by melatonin. An aberrant increase in mitochondrial respiration was also significantly suppressed by melatonin, indicating that melatonin ameliorates the mitochondrial dysfunction induced by DOX treatment. The treatment of A549 cells with luzindole, a potent inhibitor of melatonin receptors, failed to prevent the effects of melatonin treatment on mitochondrial functions and premature senescence in cells also treated with DOX; this suggests that melatonin suppresses DOX-induced senescence in a melatonin receptor-independent manner. Together, these results reveal that melatonin has an inhibitory effect of melatonin on premature senescence at the cellular level and that melatonin protects A549 cells from DOX-induced senescence. Thus, melatonin might have the therapeutic potential to prevent the side effects of anticancer drug therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cellular Senescence/drug effects , Doxorubicin/pharmacology , Lung Neoplasms/pathology , Melatonin/pharmacology , Mitochondria/drug effects , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin B/metabolism , Cyclin-Dependent Kinases , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Time Factors , Tryptamines/pharmacology
12.
J Physiol Pharmacol ; 63(6): 641-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23388480

ABSTRACT

Melatonin (MT) is a neurohormone synthesized and secreted by the pineal gland. MT plays an important role in the regulation of physiological and neuroendocrine functions. The purpose of this study was to assess the overall effect of melatonin on neuropathic pain, the type of melatonin receptor involved, and potential role of the opioid system and GABA(A) receptors. The experiments were conducted by using the animal neuropathic pain model (CCI). The rats with CCI showed the characteristic for the mechanical allodynia and thermal hyperalgesia signs that were calculated by using the von Frey's and Hargreaves' tests. The conducted studies measured the effects of intraperitoneal administration of naloxone (opioid antagonist), prazosin (MT3 antagonist), luzindole (MT1/MT2 receptor antagonist), picrotoxin (GABA(A) antagonist) and flumazenil (benzodiazepine antagonist) on the antinociceptive effects caused by melatonin. Melatonin caused the increase in the pain threshold of the mechanical allodynia and the slight increase in the threshold of the thermal hyperalgesia. The pre-treatment with naloxone completely abolished the antinociceptive effects of melatonin in von Frey's test, but not thermal sensation in the Hargreaves's test. Prazosin did not have any effects, while administration of luzindole significantly suppressed the antinociceptive effect of melatonin. The antiallodynic effect of MT was also abolished by flumazenil and picrotoxin. Melatonin influences the mechanical allodynia but not thermal hyperalgesia via activation of opioid system and benzodiazepine-GABAergic pathway. Antinociceptive effects of melatonin are mostly related to the MT1/MT2 receptors interaction.


Subject(s)
Analgesics/pharmacology , Hyperalgesia/drug therapy , Melatonin/pharmacology , Receptors, GABA-A/drug effects , Receptors, Opioid/drug effects , Sciatic Neuropathy/drug therapy , Sciatica/drug therapy , Analgesics/administration & dosage , Animals , Behavior, Animal/drug effects , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Disease Models, Animal , GABA-A Receptor Antagonists/pharmacology , Hot Temperature , Hyperalgesia/diagnosis , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Melatonin/administration & dosage , Narcotic Antagonists/pharmacology , Pain Measurement , Pain Perception/drug effects , Pain Threshold/drug effects , Pressure , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Receptors, Opioid/metabolism , Sciatic Neuropathy/diagnosis , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Sciatica/diagnosis , Sciatica/metabolism , Sciatica/physiopathology , Time Factors
13.
Article in English | MEDLINE | ID: mdl-22200560

ABSTRACT

Melatonin has both neuritogenic and neuroprotective effects in mammalian cell lines such as neuroblastoma cells. The mechanisms of action include receptor-coupled processes, direct binding and modulation of calmodulin and protein kinase C, and direct scavenging of free radicals. While melatonin is produced in invertebrates and has influences on their physiology and behavior, little is known about its mechanisms of action. We studied the influence of melatonin on neuritogenesis in well-differentiated, extensively-arborized crustacean x-organ neurosecretory neurons. Melatonin significantly increased neurite area in the first 24h of culture. The more physiological concentrations, 1 nM and 1 pM, increased area at 48 h also, whereas the pharmacological 1 µM concentration appeared to have desensitizing effects by this time. Luzindole, a vertebrate melatonin receptor antagonist, had surprising and significant agonist-like effects in these invertebrate cells. Melatonin receptors have not yet been studied in invertebrates. However, the presence of membrane-bound receptors in this population of crustacean neurons is indicated by this study. Melatonin also has significant neuroprotective effects, reversing the inhibition of neuritogenesis by 200 and 500 µM hydrogen peroxide. Because this is at least in part a direct action not requiring a receptor, melatonin's protection from oxidative stress is not surprisingly phylogenetically-conserved.


Subject(s)
Brachyura/drug effects , Melatonin/pharmacology , Neuroprotective Agents/pharmacology , Neurosecretory Systems/drug effects , Animals , Arthropod Proteins/metabolism , Brachyura/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , Melatonin/metabolism , Neurites/drug effects , Neurites/metabolism , Neuroprotective Agents/metabolism , Neurosecretory Systems/cytology , Neurosecretory Systems/metabolism , Oxidative Stress/drug effects , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Time Factors , Tryptamines/pharmacology
14.
Eur J Med Chem ; 46(9): 4252-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21764185

ABSTRACT

A novel class of imidazo[1,2-a]pyridines as melatonin receptor ligands is designed and synthesized. The affinities of 3-(6-methoxy-2-phenylimidazo[1,2-a]pyridine-3-yl)-N-methyl-propionamide 8, N-[2-(6-methoxy-2-phenylimidazo[1,2-a]pyridine-3-yl)-ethyl]-acetamide 13 and N-(1-hydroxy-3-(5-methoxy-2-phenyl-1H-indol-3-yl)propan-2-yl)acetamide 18 are evaluated for binding on melatonin receptors. Compound 8 present good selectivity for MT(2) over MT(1) (MT(1)/MT(2) = 19) and compound 13 have good affinities for both MT(1) (Ki :28 nM) and MT(2) (Ki : 8 nM).


Subject(s)
Pyridines/chemical synthesis , Pyridines/pharmacology , Receptors, Melatonin/drug effects , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Drug Design , Drug Evaluation, Preclinical , Humans , Ligands , Magnetic Resonance Spectroscopy , Pyridines/chemistry , Pyridines/metabolism , Receptors, Melatonin/metabolism , Spectrophotometry, Infrared
15.
Respir Physiol Neurobiol ; 175(2): 212-9, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21084061

ABSTRACT

Seven-transmembrane receptors mediate diverse skeletal muscle responses for a wide variety of stimuli, via activation of heterotrimeric G-proteins. Herein we evaluate the expression and activation of rat diaphragm or cultured skeletal muscle G-proteins using [(35)S]GTPγS. Total membrane Gα subunit content was 4-7 times higher in rat primary cultured myotubes and L6 cell line than in diaphragm (32.6±1.2fmol/mg protein) and 7-27% of them were in the active conformational state. Immunoprecipitation assay showed equal expression of diaphragm Gαs, Gαq and Gαi/o. Addition of GDP allowed the measurement of G-protein activation by different GPCR, including adrenoceptor, adenosine, melatonin and muscarinic receptors. Diaphragm denervation resulted in a marked increase in both total and active state G-protein levels. Together, the results show that [(35)S]GTPγS binding assay is a sensitive and valuable method to evaluate GPCR activity in skeletal muscle cells, which is of particular interest for pharmacological analysis of drugs with potential use in the management of respiratory muscle failure.


Subject(s)
Diaphragm/enzymology , Heterotrimeric GTP-Binding Proteins/physiology , Animals , Cells, Cultured , Diaphragm/drug effects , Diaphragm/innervation , Guanosine Diphosphate/pharmacology , Male , Muscarinic Agonists/pharmacology , Oxotremorine/pharmacology , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Rats , Rats, Wistar , Receptors, Adrenergic/drug effects , Receptors, Adrenergic/physiology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/physiology , Receptors, Melatonin/drug effects , Receptors, Melatonin/physiology , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/physiology , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/physiology
16.
World J Biol Psychiatry ; 11(2): 148-53, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20109111

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the acute modulation of the neurotrophin Brain-derived neurotrophic factor (BDNF) by the novel antidepressant agomelatine and the relative contribution of its melatonergic and serotonergic receptor components. METHODS: BDNF mRNA levels were measured in rat hippocampus and prefrontal cortex after acute administration of agomelatine, melatonin or the 5-HT(2C) antagonist S32006. RESULTS: BDNF expression was significantly increased 16 h after acute agomelatine administration, an effect that follows a specific temporal profile, is limited to the prefrontal cortex and it is due to changes of specific neurotrophin transcripts. Moreover, the acute up-regulation of BDNF mRNA levels appears to be the result of a synergistic effect between the melatonergic properties of agomelatine as MT1/MT2 agonist and its serotonergic 5-HT(2C) antagonism, since either melatonin or the 5-HT(2C) antagonist S32006 does not mimic the effects of agomelatine. CONCLUSIONS: These data provide evidence that acute agomelatine treatment modulates the expression of BDNF through a functional interaction between melatonergic MT1/MT2 and serotonergic 5-HT(2C) receptors, supporting the notion that intracellular events can be regulated via a synergistic activity of different neuromodulatory systems.


Subject(s)
Acetamides/pharmacology , Brain-Derived Neurotrophic Factor/analysis , Hypnotics and Sedatives/pharmacology , Prefrontal Cortex/chemistry , Animals , Gene Expression/drug effects , Hippocampus/chemistry , Hippocampus/drug effects , Indoles/pharmacology , Male , Melatonin/pharmacology , Prefrontal Cortex/drug effects , Pyridines/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Melatonin/drug effects , Receptors, Melatonin/physiology , Receptors, Serotonin, 5-HT2/drug effects , Receptors, Serotonin, 5-HT2/physiology , Serotonin Antagonists/pharmacology , Up-Regulation/drug effects
17.
Neuropsychopharmacol Hung ; 12(4): 469-75, 2010 Dec.
Article in Hungarian | MEDLINE | ID: mdl-21220792

ABSTRACT

SSRI antiepressants have been widely used for treating depressive symptoms for more than two decades. Despite their frequent usage, meta-analyses proved that only 20-25% of the patients had achieved long term remission. The introduction and spreading of dual-acting agents increased remission rate, but many of the patients with depressive symptoms still suffer from the disorder due to partial pharmacotherapeutic efficacy. Chronobiological disturbances might play an important role both in the pathophysiology and in the ongoing symptoms of depression. Pathological alterations in the melatonergic system may act as the first, obscure signs of the onset of depression. Agomelatine, a new antidepressive agent may offer new possibilities in the pharmacotherapy of depression, due to its synergistic melatonergic-serotonergic activity.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Circadian Rhythm , Depression/drug therapy , Depressive Disorder/drug therapy , Hypnotics and Sedatives/pharmacology , Melatonin/metabolism , Serotonin/metabolism , Sleep Disorders, Circadian Rhythm/drug therapy , Acetamides/therapeutic use , Antidepressive Agents/therapeutic use , Depression/metabolism , Depressive Disorder/metabolism , Drug Synergism , Humans , Hypnotics and Sedatives/therapeutic use , Melatonin/agonists , Melatonin/pharmacology , Meta-Analysis as Topic , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Serotonin/pharmacology , Sleep Disorders, Circadian Rhythm/metabolism , Sleep Disorders, Circadian Rhythm/physiopathology
18.
Prescrire Int ; 18(104): 241-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20020562

ABSTRACT

(1) When an antidepressant is considered a necessary addition to psychological support in treating patients with depression, the first-line drug is a tricyclic such as clomipramine or a selective serotonin reuptake inhibitor (SSRI) such as paroxetine; (2) Agomelatine, a melatonin receptor agonist, is approved in the European Union for the treatment of depression; (3) Available evaluation does not include any clinical trials designed to compare the efficacy of agomelatine with that of a tricyclic or a selective serotonin reuptake inhibitor. Most data come from 7 placebo-controlled trials; (4) Agomelatine (25 mg/day) was statistically more effective (on a rating scale) than placebo in only 3 of these 7 trials. The clinical relevance of the score improvements is questionable. No data are available on the cure rate or on suicide prevention; (5) In one trial, increasing the daily dose from 25 mg to 50 mg provided no supplementary benefit; (6) A trial in 367 patients failed to show that agomelatine was any more effective than placebo in preventing new depressive episodes (29% after one year). In another trial including 339 patients, the relapse rate was statistically lower after 6 months on agomelatine (20.6%) than on placebo (41.4%); (7) Very high doeses of agomelatine are oncogenic in animals. The risk in humans is not known. Dizziness, gastrointestinal and cutaneous disorders have been observed. Agomelatine is probably hepatotoxic; (8) In summary, agomelatine has unproven efficacy and poorly documented adverse effects. It is better to continue to use older antidepressants such as tricyclics or serotonin reuptake inhibitors.


Subject(s)
Acetamides/therapeutic use , Antidepressive Agents/therapeutic use , Depression/drug therapy , Depressive Disorder/drug therapy , Hypnotics and Sedatives/therapeutic use , Melatonin/agonists , Receptors, Melatonin/drug effects , Acetamides/administration & dosage , Acetamides/adverse effects , Acetamides/pharmacology , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacology , Bias , Carcinogens , Clinical Trials as Topic , Cyclohexanols/administration & dosage , Cyclohexanols/adverse effects , Cyclohexanols/pharmacology , Cyclohexanols/therapeutic use , Double-Blind Method , Drug Approval , Europe , Fluoxetine/administration & dosage , Fluoxetine/adverse effects , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Hypnotics and Sedatives/pharmacology , Melatonin/pharmacology , Melatonin/therapeutic use , Paroxetine/administration & dosage , Paroxetine/adverse effects , Paroxetine/pharmacology , Paroxetine/therapeutic use , Psychiatric Status Rating Scales , Recurrence , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Suicide , Treatment Outcome , Venlafaxine Hydrochloride
19.
Hypertens Res ; 32(4): 242-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19262491

ABSTRACT

Melatonin plays a role in blood pressure (BP) control. The aim of this study was to determine whether melatonin concentrations and melatonin receptor levels are altered in L-NAME-treated, NO-deficient hypertensive rats. Two groups of male adult Wistar rats were investigated: rats (n=36) treated with NO-synthase inhibitor L-NAME (40 mg kg(-1)) and age-matched controls (n=36). BP was measured weekly by tail-cuff plethysmography. After 4 weeks, L-NAME administration increased BP (178+/-1 vs. control 118+/-1 mm Hg). At the end of treatment, rats were killed in regular 4 h intervals over a 24-h period. Melatonin concentrations in the plasma, pineal gland, heart and kidney and melatonin receptor (MT(1)) density in the aorta were determined. A significant daily rhythm of melatonin concentrations was found in the blood, pineal gland, kidney and heart of both control and hypertensive rats. Peak nighttime pineal melatonin concentrations were higher in L-NAME-treated rats than in controls (3.38+/-0.48 vs. 1.75+/-0.33 ng per pineal gland). No differences between both groups were found in melatonin concentrations in blood, kidney and heart or in the MT(1) receptor density in the aorta. Our results suggest that L-NAME treatment enhances melatonin production in the pineal gland, potentially by decreasing an inhibitory effect of NO on melatonin production in the pineal gland. However, the enhanced pineal melatonin formation was insufficient to increase melatonin concentrations in circulation, heart and kidney of L-NAME-treated rats, indicating an increased use of melatonin in hypertensive animals.


Subject(s)
Enzyme Inhibitors/toxicity , Hypertension/metabolism , Melatonin/metabolism , NG-Nitroarginine Methyl Ester/toxicity , Nitric Oxide Synthase/antagonists & inhibitors , Pineal Gland/metabolism , Receptors, Melatonin/metabolism , Animals , Aorta/pathology , Blotting, Western , Circadian Rhythm/physiology , Hypertension/chemically induced , Kidney/pathology , Light , Male , Myocardium/pathology , Pineal Gland/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radioimmunoassay , Rats , Rats, Wistar , Receptors, Melatonin/biosynthesis , Receptors, Melatonin/drug effects , Reverse Transcriptase Polymerase Chain Reaction
20.
Psychiatry Res ; 165(3): 201-14, 2009 Feb 28.
Article in English | MEDLINE | ID: mdl-19181389

ABSTRACT

Profound disturbances in sleep architecture occur in major depressive disorders (MDD) and in bipolar affective disorders. Reduction in slow wave sleep, decreased latency of rapid eye movement (REM) sleep and abnormalities in the timing of REM/non-REM sleep cycles have all been documented in patients with MDD. It is thus evident that an understanding of the basic mechanisms of sleep regulation is essential for an analysis of the pathophysiology of depressive disorders. The suprachiasmatic nucleus (SCN), which functions as the body's master circadian clock, plays a major role in the regulation of the sleep/wakefulness rhythm and interacts actively with the homeostatic processes that regulate sleep. The control of melatonin secretion by the SCN, the occurrence of high concentrations of melatonin receptors in the SCN, and the suppression of electrical activity in the SCN by melatonin all underscore the major influence which this neurohormone has in regulating the sleep/wake cycle. The transition from wakefulness to high sleep propensity is associated with the nocturnal rise of endogenous melatonin secretion. Various lines of evidence show that depressed patients exhibit disturbances in both the amplitude and shape of the melatonin secretion rhythm and that melatonin can improve the quality of sleep in these patients. The choice of a suitable antidepressant that improves sleep quality is thus important while treating a depressive disorder. The novel antidepressant agomelatine, which combines the properties of a 5-HT(2C) antagonist and a melatonergic MT(1)/MT(2) receptor agonist, has been found very effective for resetting the disturbed sleep/wake cycle and in improving the clinical status of MDD. Agomelatine has also been found useful in treating sleep problems and improving the clinical status of patients suffering from seasonal affective disorder.


Subject(s)
Depression/epidemiology , Depression/physiopathology , Melatonin/metabolism , Sleep Stages , Sleep Wake Disorders/epidemiology , Sleep Wake Disorders/metabolism , Acetamides/pharmacology , Acetamides/therapeutic use , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Circadian Rhythm , Depression/drug therapy , Humans , Prevalence , Receptors, Melatonin/drug effects , Sleep/physiology , Suprachiasmatic Nucleus/metabolism , Wakefulness/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...