Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Transl Psychiatry ; 13(1): 54, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36788216

ABSTRACT

Morphological alterations of the endosomal compartment have been widely described in post-mortem brains from Alzheimer's disease (AD) patients and subjects with Down syndrome (DS) who are at high risk for AD. Immunostaining with antibodies against endosomal markers such as Early Endosome Antigen 1 (EEA1) revealed increased size of EEA1-positive puncta. In DS, peripheral cells such as peripheral blood mononuclear cells (PBMCs) and fibroblasts, share similar phenotype even in the absence of AD. We previously found that PBMCs from AD patients have larger EEA1-positive puncta, correlating with brain amyloid load. Here we analysed the endosomal compartment of fibroblasts from a very well characterised cohort of AD patients (IMABio3) who underwent thorough clinical, imaging and biomarkers assessments. Twenty-one subjects were included (7 AD with mild cognitive impairment (AD-MCI), 7 AD with dementia (AD-D) and 7 controls) who had amyloid-PET at baseline (PiB) and neuropsychological tests at baseline and close to skin biopsy. Fibroblasts isolated from skin biopsies were immunostained with anti-EEA1 antibody and imaged using a spinning disk microscope. Endosomal compartment ultrastructure was also analysed by electron microscopy. All fibroblast lines were genotyped and their AD risk factors identified. Our results show a trend to an increased EEA1-positive puncta volume in fibroblasts from AD-D as compared to controls (p.adj = 0.12) and reveal enhanced endosome area in fibroblasts from AD-MCI and AD-AD versus controls. Larger puncta size correlated with PiB retention in different brain areas and with worse cognitive scores at the time of biopsy as well as faster decline from baseline to the time of biopsy. Finally, we identified three genetic risk factors for AD (ABCA1, COX7C and MYO15A) that were associated with larger EEA1 puncta volume. In conclusion, the endosomal compartment in fibroblasts could be used as cellular peripheral biomarker for both amyloid deposition and cognitive decline in AD patients.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Amyloid , Amyloid beta-Peptides , Endosomes/pathology , Fibroblasts , Leukocytes, Mononuclear , Positron-Emission Tomography
2.
Front Pharmacol ; 9: 587, 2018.
Article in English | MEDLINE | ID: mdl-29930510

ABSTRACT

Previous data showed that neuropathic pain induced by mechanical lesion of peripheral nerves has specific characteristics and responds differently to alleviating drugs at cephalic versus extracephalic level. This is especially true for tricyclic antidepressants currently used for alleviating neuropathic pain in humans which are less effective against cephalic neuropathic pain. Whether this also applies to the antidepressant agomelatine, with its unique pharmacological properties as MT1/MT2 melatonin receptor agonist and 5-HT2B/5-HT2C serotonin receptor antagonist, has been investigated in two rat models of neuropathic pain. Acute treatments were performed 2 weeks after unilateral chronic constriction (ligation) injury to the sciatic nerve (CCI-SN) or the infraorbital nerve (CCI-ION), when maximal mechanical allodynia had developed in ipsilateral hindpaw or vibrissal pad, respectively, in Sprague-Dawley male rats. Although agomelatine (45 mg/kg i.p.) alone was inactive, co-treatment with gabapentin, at an essentially ineffective dose (50 mg/kg i.p.) on its own, produced marked anti-allodynic effects, especially in CCI-ION rats. In both CCI-SN and CCI-ION models, suppression of mechanical allodynia by 'agomelatine + gabapentin' could be partially mimicked by the combination of 5-HT2C antagonist (SB 242084) + gabapentin, but not by melatonin or 5-HT2B antagonist (RS 127445, LY 266097), alone or combined with gabapentin. In contrast, pretreatment by idazoxan, propranolol or the ß2 antagonist ICI 118551 markedly inhibited the anti-allodynic effect of 'agomelatine + gabapentin' in both CCI-SN and CCI-ION rats, whereas pretreatment by the MT1/MT2 receptor antagonist S22153 was inactive. Altogether these data indicate that 'agomelatine + gabapentin' is a potent anti-allodynic combination at both cephalic and extra-cephalic levels, whose action implicates α2- and ß2-adrenoreceptor-mediated noradrenergic neurotransmission.

3.
Sci Rep ; 8: 45907, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374847

ABSTRACT

Chronic stress is known to induce not only anxiety and depressive-like phenotypes in mice but also cognitive impairments, for which the action of classical antidepressant compounds remains unsatisfactory. In this context, we investigated the effects of chronic social defeat stress (CSDS) on anxiety-, social- and cognitive-related behaviors, as well as hippocampal Bdnf, synaptic plasticity markers (PSD-95, Synaptophysin, Spinophilin, Synapsin I and MAP-2), and epigenetic modifying enzymes (MYST2, HDAC2, HDAC6, MLL3, KDM5B, DNMT3B, GADD45B) gene expression in C57BL/6J mice. CSDS for 10 days provoked long-lasting anxious-like phenotype in the open field and episodic memory deficits in the novel object recognition test. While total Bdnf mRNA level was unchanged, Bdnf exon IV, MAP-2, HDAC2, HDAC6 and MLL3 gene expression was significantly decreased in the CSDS mouse hippocampus. In CSDS mice treated 3 weeks with 50 mg/kg/d agomelatine, an antidepressant with melatonergic receptor agonist and 5-HT2C receptor antagonist properties, the anxious-like phenotype was not reversed, but the treatment successfully prevented the cognitive impairments and hippocampal gene expression modifications. Altogether, these data evidenced that, in mice, agomelatine was effective in alleviating stress-induced altered cognitive functions, possibly through a mechanism involving BDNF signaling, synaptic plasticity and epigenetic remodeling.


Subject(s)
Acetamides/administration & dosage , Cognitive Dysfunction/drug therapy , Memory Disorders/drug therapy , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/administration & dosage , Brain-Derived Neurotrophic Factor/genetics , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Depressive Disorder/drug therapy , Depressive Disorder/genetics , Depressive Disorder/pathology , Disease Models, Animal , Disks Large Homolog 4 Protein/genetics , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Stress, Psychological/genetics , Stress, Psychological/pathology , Synapsins/genetics , Synaptophysin/genetics
4.
Pain ; 158(1): 149-160, 2017 01.
Article in English | MEDLINE | ID: mdl-27984527

ABSTRACT

Antidepressants are first-line treatments of neuropathic pain but not all these drugs are really effective. Agomelatine is an antidepressant with a novel mode of action, acting as an MT1/MT2 melatonergic receptor agonist and a 5-HT2C receptor antagonist that involves indirect norepinephrine release. Melatonin, serotonin, and norepinephrine have been involved in the pathophysiology of neuropathic pain. Yet, no study has been conducted to determine agomelatine effects on neuropathic pain in animal models. Using 3 rat models of neuropathic pain of toxic (oxaliplatin/OXA), metabolic (streptozocin/STZ), and traumatic (sciatic nerve ligation/CCI [chronic constriction nerve injury]) etiologies, we investigated the antihypersensitivity effect of acute and repeated agomelatine administration. We then determined the influence of melatonergic, 5-HT2C, α-2 and ß-1/2 adrenergic receptor antagonists in the antihypersensitivity effect of agomelatine. The effect of the combination of agomelatine + gabapentin was evaluated using an isobolographic approach. In STZ and CCI models, single doses of agomelatine significantly and dose dependently reduced mechanical hypersensitivity. After daily administrations for 2 weeks, this effect was confirmed in the CCI model and agomelatine also displayed a marked antihypersensitivity effect in the OXA model. The antihypersensitivity effect of agomelatine involved melatonergic, 5-HT2C, and α-2 adrenergic receptors but not beta adrenoceptors. The isobolographic analysis demonstrated that the combination of agomelatine + gabapentin had additive effects. Agomelatine exerts a clear-cut antihypersensitivity effect in 3 different neuropathic pain models. Its effect is mediated by melatonergic and 5-HT2C receptors and, although agomelatine has no affinity, also by α-2 adrenergic receptors. Finally, agomelatine combined with gabapentin produces an additive antihypersensitivity effect.


Subject(s)
Acetamides/therapeutic use , Hypnotics and Sedatives/therapeutic use , Neuralgia/drug therapy , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Amines/therapeutic use , Animals , Antineoplastic Agents/toxicity , Constriction, Pathologic/complications , Cyclohexanecarboxylic Acids/therapeutic use , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Gabapentin , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Idazoxan/therapeutic use , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuralgia/etiology , Organoplatinum Compounds/toxicity , Oxaliplatin , Pain Measurement , Rats , Rats, Sprague-Dawley , Thiophenes/therapeutic use , gamma-Aminobutyric Acid/therapeutic use
5.
Eur Neuropsychopharmacol ; 26(9): 1448-1464, 2016 09.
Article in English | MEDLINE | ID: mdl-27422761

ABSTRACT

A major domain of depression is decreased motivation for reward. Translational automated tests can be applied in humans and animals to study operant reward behaviour, aetio-pathophysiology underlying deficits therein, and effects of antidepressant treatment. Three inter-related experiments were conducted to investigate depression-relevant effects of chronic psychosocial stress on operant behaviour in mice. (A) Non-manipulated mice were trained on a complex reversal learning (CRL) test with sucrose reinforcement; relative to vehicle (VEH), acute antidepressant agomelatine (AGO, 25mg/kg p.o.) increased reversals. (B) Mice underwent chronic social defeat (CSD) or control handling (CON) on days 1-15, and were administered AGO or VEH on days 10-22. In a progressive ratio schedule motivation test for sucrose on day 15, CSD mice made fewer responses; AGO tended to reverse this effect. In a CRL test on day 22, CSD mice completed fewer reversals; AGO tended to increase reversals in CSD mice associated with an adaptive increase in perseveration. (C) Mice with continuous operant access to water and saccharin solution in the home cage were exposed to CSD or CON; CSD mice made fewer responses for saccharin and water and drank less saccharin in the active period, and drank more water in the inactive period. In a separate CSD cohort, repeated AGO was without effect on these home cage operant and consummatory changes. Overall, this study demonstrates that psychosocial stress in mice leads to depression-relevant decreases in motivation and cognition in operant reward tests; partial reversal of these deficits by AGO provides evidence for predictive validity.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Conditioning, Operant/drug effects , Motivation/drug effects , Reward , Stress, Psychological/drug therapy , Acetamides/pharmacokinetics , Animals , Antidepressive Agents/pharmacokinetics , Automation, Laboratory , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Dietary Sucrose , Dominance-Subordination , Feeding Behavior/drug effects , Feeding Behavior/physiology , Indoles/pharmacology , Male , Melatonin/pharmacology , Mice, Inbred C57BL , Models, Animal , Pyridines/pharmacology , Receptor, Serotonin, 5-HT2C/metabolism , Reversal Learning/drug effects , Saccharin , Serotonin Antagonists/pharmacology
6.
J Psychopharmacol ; 29(10): 1119-28, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26268533

ABSTRACT

The present double-blind, placebo-controlled study evaluates the effects of agomelatine and the selective serotonin reuptake inhibitor escitalopram on sexual dysfunction in healthy men and women. METHODS: A total of 133 healthy volunteers (67 men, 66 women) were randomly assigned to agomelatine (25 or 50 mg) or escitalopram (20 mg) or placebo for nine weeks. Sexual acceptability was evaluated by using the psychotropic-related sexual dysfunction questionnaire 5-items total score and sexual dysfunction relative to each sub-score (in 110 volunteers with sexual activity). Sexual dysfunction was evaluated at baseline and after two, five and eight weeks of treatment and one week after drug discontinuation. RESULTS: The psychotropic-related sexual dysfunction questionnaire 5-items total score was significantly lower in both agomelatine groups versus escitalopram at all visits (p < 0.01 to p < 0.0001) with no difference between agomelatine and placebo nor between both agomelatine doses. Similar results were observed after drug discontinuation. The total score was significantly higher in the escitalopram group than in the placebo group at each post-baseline visit (p < 0.01 to p < 0.001). Similar results were observed regardless of volunteers' gender. Compared to placebo, only escitalopram significantly impaired dysfunction relative to "delayed orgasm or ejaculation" (p < 0.01) and "absence of orgasm or ejaculation" (p < 0.05 to p < 0.01). The percentage of participants with a sexual dysfunction was higher in the escitalopram group than in agomelatine groups (p < 0.01 to p < 0.05) and placebo (p < 0.01). CONCLUSION: The study confirms the better sexual acceptability profile of agomelatine (25 or 50 mg) in healthy men and women, compared to escitalopram. TRIAL REGISTRATION NAME: Evaluation of the effect of agomelatine and escitalopram on emotions and motivation in healthy male and female volunteers. TRIAL REGISTRATION NUMBER: ISRCTN75872983.


Subject(s)
Acetamides/administration & dosage , Citalopram/administration & dosage , Hypnotics and Sedatives/administration & dosage , Selective Serotonin Reuptake Inhibitors/administration & dosage , Sexual Dysfunctions, Psychological/drug therapy , Adult , Double-Blind Method , Emotions/drug effects , Female , Healthy Volunteers , Humans , Male , Psychiatric Status Rating Scales , Surveys and Questionnaires , Young Adult
7.
J Neurosci ; 34(6): 2015-24, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24501344

ABSTRACT

Abnormalities of synaptic transmission in the hippocampus represent an integral part of the altered programming triggered by early life stress, which enhances the vulnerability to stress-related disorders in the adult life. Rats exposed to prenatal restraint stress (PRS) develop enduring biochemical and behavioral changes characteristic of an anxious/depressive-like phenotype. Most neurochemical abnormalities in PRS rats are found in the ventral hippocampus, a region that encodes memories related to stress and emotions. We have recently demonstrated a causal link between the reduction of glutamate release in the ventral hippocampus and anxiety-like behavior in PRS rats. To confer pharmacological validity to the glutamatergic hypothesis of stress-related disorders, we examined whether chronic treatment with two antidepressants with different mechanisms of action could correct the defect in glutamate release and associated behavioral abnormalities in PRS rats. Adult unstressed or PRS rats were treated daily with either agomelatine (40 mg/kg, i.p.) or fluoxetine (5 mg/kg, i.p.) for 21 d. Both treatments reversed the reduction in depolarization-evoked glutamate release and in the expression of synaptic vesicle-associated proteins in the ventral hippocampus of PRS rats. Antidepressant treatment also corrected abnormalities in anxiety-/depression-like behavior and social memory performance in PRS rats. The effect on glutamate release was strongly correlated with the improvement of anxiety-like behavior and social memory. These data offer the pharmacological demonstration that glutamatergic hypofunction in the ventral hippocampus lies at the core of the pathological phenotype caused by early life stress and represents an attractive pharmacological target for novel therapeutic strategies.


Subject(s)
Antidepressive Agents/therapeutic use , Glutamic Acid/metabolism , Prenatal Exposure Delayed Effects/drug therapy , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Animals , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/psychology , Depression/drug therapy , Depression/metabolism , Depression/psychology , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Sprague-Dawley , Stress, Psychological/psychology , Treatment Outcome
8.
BMC Neurosci ; 14: 75, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23895555

ABSTRACT

BACKGROUND: Growing compelling evidence from clinical and preclinical studies has demonstrated the primary role of alterations of glutamatergic transmission in cortical and limbic areas in the pathophysiology of mood disorders. Chronic antidepressants have been shown to dampen endogenous glutamate release from rat hippocampal synaptic terminals and to prevent the marked increase of glutamate overflow induced by acute behavioral stress in frontal/prefrontal cortex. Agomelatine, a new antidepressant endowed with MT1/MT2 agonist and 5-HT2C serotonergic antagonist properties, has shown efficacy at both preclinical and clinical levels. RESULTS: Chronic treatment with agomelatine, or with the reference drug venlafaxine, induced a marked decrease of depolarization-evoked endogenous glutamate release from purified hippocampal synaptic terminals in superfusion. No changes were observed in GABA release. This effect was accompanied by reduced accumulation of SNARE protein complexes, the key molecular effector of vesicle docking, priming and fusion at presynaptic membranes. CONCLUSIONS: Our data suggest that the novel antidepressant agomelatine share with other classes of antidepressants the ability to modulate glutamatergic transmission in hippocampus. Its action seems to be mediated by molecular mechanisms located on the presynaptic membrane and related with the size of the vesicle pool ready for release.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Cyclohexanols/pharmacology , Glutamic Acid/metabolism , Hippocampus/cytology , Synaptosomes/drug effects , Analysis of Variance , Animals , Calcium Ionophores/pharmacology , Ionomycin/pharmacology , Male , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , SNARE Proteins/metabolism , Syntaxin 1/metabolism , Venlafaxine Hydrochloride , gamma-Aminobutyric Acid/metabolism
9.
Int J Neuropsychopharmacol ; 16(2): 323-38, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22310059

ABSTRACT

Agomelatine is a novel antidepressant acting as an MT1/MT2 melatonin receptor agonist/5-HT2C serotonin receptor antagonist. Because of its peculiar pharmacological profile, this drug caters the potential to correct the abnormalities of circadian rhythms associated with mood disorders, including abnormalities of the sleep/wake cycle. Here, we examined the effect of chronic agomelatine treatment on sleep architecture and circadian rhythms of motor activity using the rat model of prenatal restraint stress (PRS) as a putative 'aetiological' model of depression. PRS was delivered to the mothers during the last 10 d of pregnancy. The adult progeny ('PRS rats') showed a reduced duration of slow wave sleep, an increased duration of rapid eye movement (REM) sleep, an increased number of REM sleep events and an increase in motor activity before the beginning of the dark phase of the light/dark cycle. In addition, adult PRS rats showed an increased expression of the transcript of the primary response gene, c-Fos, in the hippocampus just prior to the beginning of the dark phase. All these changes were reversed by a chronic oral treatment with agomelatine (2000 ppm in the diet). The effect of agomelatine on sleep was largely attenuated by treatment with the MT1/MT2 melatonin receptor antagonist, S22153, which caused PRS-like sleep disturbances on its own. These data provide the first evidence that agomelatine corrects sleep architecture and restores circadian homeostasis in a preclinical model of depression and supports the value of agomelatine as a novel antidepressant that resynchronizes circadian rhythms under pathological conditions.


Subject(s)
Acetamides/therapeutic use , Chronobiology Disorders/drug therapy , Hypnotics and Sedatives/therapeutic use , Movement Disorders/drug therapy , Prenatal Exposure Delayed Effects/physiopathology , Sleep Wake Disorders/drug therapy , Analysis of Variance , Animals , Animals, Newborn , Arousal/drug effects , Autoradiography , Chronobiology Disorders/etiology , Disease Models, Animal , Drug Administration Schedule , Electroencephalography , Electromyography , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Movement Disorders/etiology , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy , Prenatal Exposure Delayed Effects/pathology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Melatonin/antagonists & inhibitors , Restraint, Physical/adverse effects , Sleep Wake Disorders/etiology , Thiophenes/pharmacology
10.
Brain Res ; 1466: 91-8, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22647752

ABSTRACT

Agomelatine is a potent melatonergic (MT1 and MT2) receptor agonist and 5HT(2C) antagonist that is an effective antidepressant in animal models of depression and in patients suffering from depression. Our recent studies revealed that acute restraint stress increases extracellular levels of glutamate and GABA and that these increases in amino acid efflux are inhibited by some but not all antidepressants. In view of the increasing evidence supporting a role of amino acids in the pathology of depression, the current study examined whether acute stress-mediated changes in glutamate and GABA neurotransmission are modulated by agomelatine. In agreement with our previous work, acute stress increases extracellular glutamate levels in the basolateral nucleus of the amygdala (BLA). Similarly, acute stress increases glutamate efflux in the central nucleus of the amygdala (CeA). In the hippocampus, acute stress increases glutamate efflux and elicits an oscillatory pattern of GABA efflux. Agomelatine administration (40mg/kg ip) prior to acute stress inhibited stress-mediated increases in glutamate efflux in the hippocampus, BLA and CeA. These results demonstrate that acute agomelatine administration effectively inhibits acute stress-mediated changes in extracellular glutamate in the rat hippocampus and amygdala. While acute stress did not modulate GABA efflux in these regions, agomelatine treatment induced an overall reduction of GABA levels in the hippocampus. These data suggest that agomelatine modulates amino acid efflux in limbic structures implicated in major depressive disorder.


Subject(s)
Acetamides/pharmacology , Amygdala/drug effects , Antidepressive Agents/pharmacology , Glutamic Acid/metabolism , Hippocampus/drug effects , Stress, Physiological/drug effects , gamma-Aminobutyric Acid/metabolism , Amygdala/metabolism , Animals , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
11.
Int J Neuropsychopharmacol ; 15(3): 321-35, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21473810

ABSTRACT

Agomelatine (S20098) is a novel antidepressant drug with melatonergic agonist and 5-HT2C receptor antagonist properties, displaying antidepressant/anxiolytic-like properties in animal models and in humans. In a depression/anxiety-like mouse model in which the response of the HPA axis is blunted, we investigated whether agomelatine could reverse behavioural deficits related to depression/anxiety compared to the classical selective serotonin reuptake inhibitor, fluoxetine. Adult mice were treated for 8 wk with either vehicle or corticosterone (35 µg/ml.d) via drinking water. During the final 4 wk, animals were treated with vehicle, agomelatine (10 or 40 mg/kg i.p.) or fluoxetine (18 mg/kg i.p.) and tested in several behavioural paradigms and also evaluated for home-cage activity. Our results showed that the depressive/anxiety-like phenotype induced by corticosterone treatment is reversed by either chronic agomelatine or fluoxetine treatment. Moreover, agomelatine increased the dark/light ratio of home-cage activity in vehicle-treated mice and reversed the alterations in this ratio induced by chronic corticosterone, suggesting a normalization of disturbed circadian rhythms. Finally, we investigated the effects of this new antidepressant on neurogenesis. Agomelatine reversed the decreased cell proliferation in the whole hippocampus in corticosterone-treated mice and increased maturation of newborn neurons in both vehicle- and corticosterone-treated mice. Overall, the present study suggests that agomelatine, with its distinct mechanism of action based on the synergy between the melatonergic agonist and 5-HT2C antagonist properties, provides a distinct antidepressant/anxiolytic spectrum including circadian rhythm normalization.


Subject(s)
Acetamides/pharmacology , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety Disorders/drug therapy , Depressive Disorder/drug therapy , Neurogenesis/drug effects , Animals , Anxiety Disorders/physiopathology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Corticosterone , Dendrites/drug effects , Dendrites/physiology , Depressive Disorder/physiopathology , Disease Models, Animal , Fluoxetine/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Serotonin 5-HT2 Receptor Antagonists/pharmacology
12.
Psychopharmacology (Berl) ; 221(3): 493-509, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22160164

ABSTRACT

RATIONALE: Agomelatine is described as a novel and clinical effective antidepressant drug with melatonergic (MT(1)/MT(2)) agonist and 5-HT(2C) receptor antagonist properties. Previous studies suggest that modulation of neuronal plasticity and microtubule dynamics may be involved in the treatment of depression. OBJECTIVE: The present study investigated the effects of agomelatine on microtubular, synaptic and brain-derived neurotrophic factor (BDNF) proteins in selected rat brain regions. METHODS: Adult male rats received agomelatine (40 mg/kg i.p.) once a day for 22 days. The pro-cognitive effect of agomelatine was tested in the novel object recognition task and antidepressant activity in the forced swimming test. Microtubule dynamics markers, microtubule-associated protein type 2 (MAP-2), phosphorylated MAP-2, synaptic markers [synaptophysin, postsynaptic density-95 (PSD-95) and spinophilin] and BDNF were measured by Western blot in the hippocampus, amygdala and prefrontal cortex (PFC). RESULTS: Agomelatine exerted pro-cognitive and antidepressant activity and induced molecular changes in the brain areas examined. Agomelatine enhanced microtubule dynamics in the hippocampus and to a higher magnitude in the amygdala. By contrast, in the PFC, a decrease in microtubule dynamics was observed. Spinophilin (dendritic spines marker) was decreased, and BDNF increased in the hippocampus. Synaptophysin (presynaptic) and spinophilin were increased in the PFC and amygdala, while PSD-95 (postsynaptic marker) was increased in the amygdala, consistent with the phenomena of synaptic remodelling. CONCLUSIONS: Agomelatine modulates cytoskeletal microtubule dynamics and synaptic markers. This may play a role in its pharmacological behavioural effects and may result from the melatonergic agonist and 5-HT(2C) antagonist properties of the compound.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Cognition/drug effects , Depression/drug therapy , Amygdala/drug effects , Amygdala/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Male , Microtubules/drug effects , Microtubules/metabolism , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Synapses/metabolism
13.
Eur J Pharmacol ; 674(1): 27-32, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22040921

ABSTRACT

The novel melatonergic agonist/5-HT(2C) antagonist agomelatine displays robust antidepressant properties in humans and is active in pre-clinical models predictive of antidepressant effects. In this study, we investigated its potential influence on the locomotor hyperactivity displayed by olfactory bulbectomised rats, a putative measure of potential antidepressant activity. In addition, we compared the actions of agomelatine to those of melatonin and S32006, a selective antagonist at 5-HT(2C) receptors. Vehicle, agomelatine (10 and 50mg/kg), melatonin (10 and 50mg/kg), S32006 (0.16mg/kg to 10mg/kg) and the prototypical tricyclic antidepressant, imipramine (10mg/kg), were administered by intraperitoneal injection for 14days to male, Sprague-Dawley sham-operated and bulbectomised rats. In agreement with previous studies, imipramine was active in the model and both the lower and higher doses of agomelatine also significantly and markedly reversed the bulbectomy-induced hyperactivity to a level comparable to that seen in sham operated animals, in which agomelatine exerted no effect. Similarly the 5-HT(2C) antagonist, S32006, dose-dependently and significantly attenuated hyperactivity of bulbectomised animals, albeit with a maximal effect somewhat less marked than that of agomelatine. On the other hand, melatonin did not affect the locomotor behaviour of bulbectomised rats. The activity of agomelatine in the model is consistent with its known antidepressant properties in the clinic.


Subject(s)
Acetamides/pharmacology , Hyperkinesis/drug therapy , Indoles/pharmacology , Melatonin/pharmacology , Olfactory Bulb/surgery , Pyridines/pharmacology , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Acetamides/therapeutic use , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Indoles/therapeutic use , Male , Melatonin/therapeutic use , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Serotonin 5-HT2 Receptor Antagonists/therapeutic use
14.
Pharmacol Biochem Behav ; 100(2): 284-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21843546

ABSTRACT

The novel antidepressant agomelatine behaves as an agonist at melatonergic MT(1) and MT(2) receptors and as an antagonist at serotonin 5-HT(2C) receptors. This study investigated the effects of agomelatine and fluoxetine in a genetic model of depression called H/Rouen mice Male and female H/Rouen (helpless line) and NH/Rouen (nonhelpless line) mice, received once daily for 3 weeks agomelatine (10 and 50 mg/kgi.p.), fluoxetine (10 mg/kgi.p.) or vehicle. Immobility duration in the tail suspension test (TST) was assessed on day 1 (D1), day 8 (D8), day 15 (D15) and day 22 (D22). Locomotor activity in a novel environment was assessed on day 18 (D18) and anhedonia (2-bottle sucrose preference test) was considered after the end of chronic treatment, from days 22 to 25. Agomelatine (50 mg/kg) significantly reduced immobility at D15 (p<0.01), and D22 (p<0.001) in treated H/Rouen mice whereas agomelatine at 10 mg/kg did not induce a statistically significant change. Fluoxetine reduced immobility at D8 (p<0.01), D15 (p<0.001) and D22 (p<0.001). Locomotor activity was unchanged in all treated groups as compared to vehicle groups. In the sucrose test, there was a significant decrease in sucrose preference in H/Rouen mice compared with NH/Rouen mice receiving vehicle. Both agomelatine doses (10 mg/kg (p=0.05) and 50 mg/kg (p<0.001) as well as fluoxetine (p<0.001) significantly increased the sucrose preference in H/Rouen mice as compared with H/Rouen mice that had received vehicle. These data indicate that the novel antidepressant agomelatine has antidepressant-like properties in H/Rouen mice, a genetic model of depression.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Fluoxetine/pharmacology , Animals , Disease Models, Animal , Locomotion , Mice
15.
J Neurosci Res ; 89(10): 1646-57, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21688292

ABSTRACT

The antidepressant agomelatine acts as a melatonergic receptor (MT(1)/MT(2)) agonist and 5-HT(2C) receptor antagonist. Agomelatine has demonstrated efficacy in treating depression, but its neurobiological effects merit further investigation. Preclinical studies reported that agomelatine enhances adult hippocampal neurogenesis and increases expression of several neuroplasticity-associated molecules. Recently, we showed that agomelatine normalizes hippocampal neuronal activity and promotes neurogenesis in the stress-compromised brain. To characterize further the effects of this antidepressant in the stressed brain, here we investigated whether it induces changes in the expression of synapsin I (SynI), a regulator of synaptic transmission and plasticity. Adult male rats were subjected to daily footshock stress and agomelatine treatment for 3 weeks. Their brains were subsequently stained for total and phosphorylated SynI. Chronic footshock and agomelatine induced region-specific changes in SynI expression. Whereas chronic stress increased total SynI expression in all layers of the medial prefrontal cortex, agomelatine treatment abolished some of these effects. Furthermore, chronic agomelatine administration decreased total SynI expression in the hippocampal subregions of both stressed and nonstressed rats. Importantly, chronic stress decreased the fraction of phosphorylated SynI in all layers of the medial prefrontal cortex as well as selectively in the outer and middle molecular layers of the hippocampal dentate gyrus. These stress effects were at least partially abolished by agomelatine. Altogether, our data show that chronic stress and agomelatine treatment induce region-specific changes in SynI expression and its phosphorylation. Moreover, agomelatine partially counteracts the stress effects on SynI, suggesting a modulation of synaptic function by this antidepressant.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Brain Chemistry/drug effects , Brain Chemistry/genetics , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Stress, Psychological/drug therapy , Synapsins/genetics , Animals , Chronic Disease , Disease Models, Animal , Male , Rats , Rats, Wistar , Stress, Psychological/metabolism , Synapsins/biosynthesis , Synapsins/metabolism
16.
Psychopharmacology (Berl) ; 217(3): 301-13, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21503609

ABSTRACT

RATIONALE AND OBJECTIVES: The rat model of prenatal restraint stress (PRS) replicates factors that are implicated in the etiology of anxious/depressive disorders. We used this model to test the therapeutic efficacy of agomelatine, a novel antidepressant that behaves as a mixed MT1/MT2 melatonin receptor agonist/5-HT(2c) serotonin receptor antagonist. RESULTS: Adult PRS rats showed behavioral, cellular, and biochemical abnormalities that were consistent with an anxious/depressive phenotype. These included an increased immobility in the forced swim test, an anxiety-like behavior in the elevated plus maze, reduced hippocampal levels of phosphorylated cAMP-responsive element binding protein (p-CREB), reduced hippocampal levels of mGlu2/3 and mGlu5 metabotropic glutamate receptors, and reduced neurogenesis in the ventral hippocampus, the specific portion of the hippocampus that encodes memories related to stress and emotions. All of these changes were reversed by a 3- or 6-week treatment with agomelatine (40-50 mg/kg, i.p., once a day). Remarkably, agomelatine had no effect in age-matched control rats, thereby behaving as a "disease-dependent" drug. CONCLUSIONS: These data indicate that agomelatine did not act on individual symptoms but corrected all aspects of the pathological epigenetic programming triggered by PRS. Our findings strongly support the antidepressant activity of agomelatine and suggest that the drug impacts mechanisms that lie at the core of anxious/depressive disorders.


Subject(s)
Acetamides/therapeutic use , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Hippocampus , Neurogenesis/drug effects , Prenatal Exposure Delayed Effects/prevention & control , Stress, Psychological/complications , Acetamides/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Anxiety/metabolism , Anxiety/pathology , Anxiety/prevention & control , CREB-Binding Protein/metabolism , Cell Proliferation/drug effects , Data Interpretation, Statistical , Depression/metabolism , Depression/pathology , Depression/prevention & control , Female , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/pathology , Immunoblotting , Male , Maze Learning/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism
17.
Psychopharmacology (Berl) ; 215(2): 267-75, 2011 May.
Article in English | MEDLINE | ID: mdl-21181122

ABSTRACT

RATIONALE: Neuronal plasticity is associated with depression, probably as a result of modified expression of proteins important for cellular resiliency. It is therefore important to establish if and how antidepressant drugs may be able to regulate these mechanisms in order to achieve relevant clinical effects. OBJECTIVE: We investigated the effects of chronic treatment with agomelatine (an MT(1)/MT(2) receptor agonist and 5-HT(2C) receptor antagonist) on the brain-derived neurotrophic factor (BDNF), fibroblast growth factor (FGF-2), and activity-regulated cytoskeleton-associated protein (Arc). METHODS: Animals were treated for 21 days with agomelatine, venlafaxine, or a vehicle and sacrificed 1 h (6 p.m.) or 16 h after the last injection (9 a.m.) to evaluate the messenger RNA (mRNA) and protein expression of these neuroplastic markers in the hippocampus and prefrontal cortex. RESULTS: Agomelatine, but not venlafaxine, produced major transcriptional changes in the hippocampus, where significant up-regulations of BDNF and FGF-2 were observed. Both drugs up-regulated the Arc transcription levels. No effects were observed in the prefrontal cortex. Instead, the levels of BDNF protein were elevated by agomelatine in both regions: the effects of the drug on mRNA levels in the hippocampus and cortex are different, while the effects on the protein seem to have the same cumulative result, suggesting different modulatory mechanisms in the two regions. CONCLUSIONS: Our data provide new information regarding the molecular mechanisms that contribute to the chronic effects of the new antidepressant agomelatine on brain function. The ability of agomelatine to modulate the expression of these neuroplastic molecules, which follows a circadian rhythm, may contribute to its antidepressant action.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Brain/drug effects , Gene Expression Regulation/drug effects , Neuronal Plasticity/drug effects , Animals , Brain/anatomy & histology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclohexanols/pharmacology , Cytosol/drug effects , Cytosol/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Neuronal Plasticity/genetics , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism , Time Factors , Venlafaxine Hydrochloride
18.
Behav Brain Res ; 218(1): 121-8, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21115070

ABSTRACT

The antidepressant agomelatine is a MT(1)/MT(2) receptor agonist and 5-HT(2C) antagonist. Its antidepressant activity is proposed to result from the synergy between these sets of receptors. Agomelatine-induced changes in the brain have been reported under basal conditions. Yet, little is known about its effects in the brain exposed to chronic stress as a risk factor for major depressive disorder. Recently, we described agomelatine-induced changes on neuronal activity and adult neurogenesis in the hippocampus of rats subjected to chronic footshock stress. In order to better characterize the actions of agomelatine in the stress-compromised brain, here we investigated its effects on hippocampal neurogenesis in the chronic mild stress (CMS) model. Adult male rats were subjected to various mild stressors for 5 weeks, and treated with agomelatine during the last 3 weeks of the stress period. The sucrose preference test was performed weekly to measure anhedonia, and the marble burying test was carried out at the end of the experiment to assess anxiety-like behavior. In our model, the CMS paradigm did not change sucrose preference; however, it increased marble burying behavior, indicating enhanced anxiety. Interestingly, this stress model differentially affected distinct stages of the neurogenesis process. Whereas CMS did not influence the rate of hippocampal cell proliferation, it significantly decreased the newborn cell survival and doublecortin expression in the dentate gyrus. Importantly, treatment with agomelatine completely normalized stress-affected cell survival and partly reversed reduced doublecortin expression. Taken together, these data show that agomelatine has beneficial effects on hippocampal neurogenesis in the CMS paradigm.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Cell Survival/drug effects , Hippocampus/drug effects , Stress, Physiological/drug effects , Stress, Psychological , Analysis of Variance , Animals , Behavior, Animal/drug effects , Bromodeoxyuridine/metabolism , Cell Count , Cell Proliferation/drug effects , Doublecortin Protein , Hippocampus/metabolism , Immunohistochemistry , Male , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar
19.
CNS Neurosci Ther ; 16(4): 195-207, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20236141

ABSTRACT

Agomelatine is a novel antidepressant which acts as a melatonergic (MT1/MT2) receptor agonist and serotonergic (5-HT2C) receptor antagonist. The antidepressant properties of agomelatine have been demonstrated in animal models as well as in clinical studies. Several preclinical studies reported agomelatine-induced effects on brain plasticity, mainly under basal conditions in healthy animals. Yet, it is important to unravel agomelatine-mediated changes in the brain affected by psychopathology or exposed to conditions that might predispose to mood disorders. Since stress is implicated in the etiology of depression, it is valid to investigate antidepressant-induced effects in animals subjected to chronic stress. In this context, we sought to determine changes in the brain after agomelatine treatment in chronically stressed rats. Adult male rats were subjected to footshock stress and agomelatine treatment for 21 consecutive days. Rats exposed to footshock showed a robust increase in adrenocorticotropic hormone (ACTH) and corticosterone. Chronic agomelatine treatment did not markedly influence this HPA-axis response. Whereas chronic exposure to daily footshock stress reduced c-Fos expression in the hippocampal dentate gyrus, agomelatine treatment reversed this effect and normalized neuronal activity to basal levels. Moreover, chronic agomelatine administration was associated with enhanced hippocampal cell proliferation and survival in stressed but not in control rats. Furthermore, agomelatine reversed the stress-induced decrease in doublecortin expression in the dentate gyrus. Taken together, these data show a beneficial action of agomelatine in the stress-compromised brain, where it restores stress-affected hippocampal neuronal activity and promotes adult hippocampal neurogenesis.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Hippocampus/pathology , Neurogenesis/drug effects , Neurons/drug effects , Stress, Psychological/physiopathology , Adrenocorticotropic Hormone/metabolism , Analysis of Variance , Animals , Antidepressive Agents/therapeutic use , Body Weight/drug effects , Bromodeoxyuridine/metabolism , Corticosterone/metabolism , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Eating/drug effects , Electroshock/adverse effects , Gene Expression Regulation/drug effects , Ki-67 Antigen/metabolism , Male , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Stress, Psychological/drug therapy , Stress, Psychological/pathology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...