Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Elisabeth Mocaër is active.

Publication


Featured researches published by Elisabeth Mocaër.


Nature Reviews Drug Discovery | 2010

Agomelatine, the first melatonergic antidepressant: discovery, characterization and development

Christian de Bodinat; Beatrice Guardiola-Lemaitre; Elisabeth Mocaër; Pierre Renard; Carmen Muńoz; Mark J. Millan

Current management of major depression, a common and debilitating disorder with a high social and personal cost, is far from satisfactory. All available antidepressants act through monoaminergic mechanisms, so there is considerable interest in novel non-monoaminergic approaches for potentially improved treatment. One such strategy involves targeting melatonergic receptors, as melatonin has a key role in synchronizing circadian rhythms, which are known to be perturbed in depressed states. This article describes the discovery and development of agomelatine, which possesses both melatonergic agonist and complementary 5-hydroxytryptamine 2C (5-HT2C) antagonist properties. Following comprehensive pharmacological evaluation and extensive clinical trials, agomelatine (Valdoxan/Thymanax; Servier) was granted marketing authorization in 2009 for the treatment of major depression in Europe, thereby becoming the first approved antidepressant to incorporate a non-monoaminergic mechanism of action.


Neuropsychopharmacology | 2003

Effect of Agomelatine in the Chronic Mild Stress Model of Depression in the Rat

Mariusz Papp; Piotr Gruca; Pierre-Alain Boyer; Elisabeth Mocaër

Chronic mild stress (CMS), a well-validated model of depression, was used to study the effects of the melatonin agonist and selective 5-HT2C antagonist agomelatine (S 20098) in comparison with melatonin, imipramine, and fluoxetine. All drugs were administered either 2 h before (evening treatment) or 2 h after (morning treatment) the dark phase of the 12-h light/dark cycle. Chronic (5 weeks) evening treatment with agomelatine or melatonin (both at 10 and 50 mg/kg i.p.) dose-dependently reversed the CMS-induced reduction in sucrose consumption. The magnitude and time course of the action of both drugs was comparable to that of imipramine and fluoxetine (both at 10 mg/kg i.p.); however, melatonin was less active than agomelatine at this dose. The effect of evening administration of agomelatine and melatonin was completely inhibited by an acute injection of the MT1/MT2 antagonist, S 22153 (20 mg/kg i.p.), while the antagonist had no effect in animals receiving fluoxetine or imipramine. When the drugs were administered in the morning, agomelatine caused effects similar to those observed after evening treatment (with onset of action faster than imipramine) but melatonin was ineffective. Moreover, melatonin antagonist, S 22153, did not modify the intakes in stressed animals receiving morning administration of agomelatine and in any other control and stressed groups tested in this study. These data demonstrate antidepressant-like activity of agomelatine in the rat CMS model of depression, which was independent of the time of drug administration. The efficacy of agomelatine is comparable to that of imipramine and fluoxetine, but greater than that of melatonin, which had no antidepressant-like activity after morning administration. While the evening efficacy of agomelatine can be related to its melatonin receptors agonistic properties, its morning activity, which was not inhibited by a melatonin antagonist, indicates that these receptors are certainly required, but not sufficient to sustain the agomelatine efficacy. It is therefore suggested that the antidepressant-like activity of agomelatine depends on some combination of its melatonin agonist and 5-HT2C antagonist properties.


Neuropsychopharmacology | 2010

Enhancement of Attentional Performance by Selective Stimulation of α4β2* nAChRs: Underlying Cholinergic Mechanisms

William M. Howe; Jinzhao Ji; Vinay Parikh; Sarah Williams; Elisabeth Mocaër; Caryn Trocme-Thibierge; Martin Sarter

Impairments in attention are a major component of the cognitive symptoms of neuropsychiatric and neurodegenerative disorders. Using an operant sustained attention task (SAT), including a distractor condition (dSAT), we assessed the putative pro-attentional effects of the selective α4β2* nicotinic acetylcholine receptor (nAChR) agonist S 38232 in comparison with the non-selective agonist nicotine. Neither drug benefited SAT performance. However, in interaction with the increased task demands implemented by distractor presentation, the selective agonist, but not nicotine, enhanced the detection of signals during the post-distractor recovery period. This effect is consistent with the hypothesis that second-long increases in cholinergic activity (‘transients’) mediate the detection of cues and that nAChR agonists augment such transients. Electrochemical recordings of prefrontal cholinergic transients evoked by S 38232 and nicotine indicated that the α4β2* nAChR agonist evoked cholinergic transients that were characterized by a faster rise time and more rapid decay than those evoked by nicotine. Blockade of the α7 nAChR ‘sharpens’ nicotine-evoked transients; therefore, we determined the effects of co-administration of nicotine and the α7 nAChR antagonist methyllycaconitine on dSAT performance. Compared with vehicle and nicotine alone, this combined treatment significantly enhanced the detection of signals. These results indicate that compared with nicotine, α4β2* nAChR agonists significantly enhance attentional performance and that the dSAT represents a useful behavioral screening tool. The combined behavioral and electrochemical evidence supports the hypothesis that nAChR agonist-evoked cholinergic transients, which are characterized by rapid rise time and fast decay, predict robust drug-induced enhancement of attentional performance.


Neuropsychopharmacology | 2009

Mechanisms Contributing to the Phase-Dependent Regulation of Neurogenesis by the Novel Antidepressant, Agomelatine, in the Adult Rat Hippocampus

Amélie Soumier; Mounira Banasr; Sylviane Lortet; Frédérique Masmejean; Nathalie Bernard; Lydia Kerkerian-Le-Goff; Cecilia Gabriel; Mark J. Millan; Elisabeth Mocaër; Annie Daszuta

Agomelatine is a novel antidepressant acting as a melatonergic receptor agonist and serotonergic (5-HT2C) receptor antagonist. In adult rats, chronic agomelatine treatment enhanced cell proliferation and neurogenesis in the ventral hippocampus (VH), a region pertinent to mood disorders. This study compared the effects of agomelatine on cell proliferation, maturation, and survival and investigated the cellular mechanisms underlying these effects. Agomelatine increased the ratio of mature vs immature neurons and enhanced neurite outgrowth of granular cells, suggesting an acceleration of maturation. The influence of agomelatine on maturation and survival was accompanied by a selective increase in the levels of BDNF (brain-derived neurotrophic factor) vs those of VEGF (vascular endothelial factor) and IGF-1 (insulin-like growth factor 1), which were not affected. Agomelatine also activated several cellular signals (extracellular signal-regulated kinase1/2, protein kinase B, and glycogen synthase kinase 3β) known to be modulated by antidepressants and implicated in the control of proliferation/survival. Furthermore, as agomelatine possesses both melatonergic agonist and serotonergic (5-HT2C) antagonist properties, we determined whether melatonin and 5-HT2C receptor antagonists similarly influence cell proliferation and survival. Only the 5-HT2C receptor antagonists, SB243,213 or S32006, but not melatonin, mimicked the effects of agomelatine on cell proliferation in VH. The promoting effect of agomelatine on survival was not reproduced by the 5-HT2C receptor antagonists or melatonin alone. However, it was blocked by a melatonin antagonist, S22153. These results show that agomelatine treatment facilitates all stages of neurogenesis and suggest that a joint effect of melatonin agonism and 5HT2C antagonism may be involved in promotion by agomelatine of survival in the hippocampus.


The International Journal of Neuropsychopharmacology | 2010

Behavioural and neuroplastic effects of the new-generation antidepressant agomelatine compared to fluoxetine in glucocorticoid receptor-impaired mice.

Eleni Païzanis; Thibault Renoir; Vincent Lelievre; Françoise Saurini; Maxette Melfort; Cecilia Gabriel; Nicholas Barden; Elisabeth Mocaër; Michel Hamon; Laurence Lanfumey

Major depression is associated with reduced hippocampal volume linked to stress and high glucocorticoid secretion. Glucocorticoid receptor-impaired (GR-i) mice, a transgenic model for affective disorders with hypothalamic-pituitary-adrenal (HPA) axis feedback control deficit, were used to assess the antidepressant-like effects of the mixed melatonin receptor agonist/5-HT(2C) receptor antagonist, agomelatine, compared to the selective 5-HT reuptake inhibitor (SSRI), fluoxetine, on hippocampal neurogenesis, GR and BDNF expression and antidepressant-responsive behaviour (tail suspension test, TST). GR-i and paired wild-type (WT) mice were given acute or chronic (21 d) treatment with these drugs. Both hippocampal cell proliferation and BDNF mRNA expression were down-regulated in GR-i mice, and these alterations were reversed by chronic agomelatine and fluoxetine treatments, whereas GR mRNA down-regulation was reversed only by agomelatine. Furthermore, chronic agomelatine, but not fluoxetine, increased survival of newly formed cells in the ventral part of the hippocampus without changing their phenotypic differentiation into neurons. In the TST, the enhanced immobility of GR-i mice was reduced to WT level by acute (but not chronic) fluoxetine and chronic (but not acute) agomelatine. These results indicate that agomelatine reversed the neuroplastic changes and helpless behaviour associated with HPA axis alterations in GR-i mice, suggesting neurobiological and behavioural effects mostly similar to those typically seen with classical antidepressants such as fluoxetine, but through clearly distinct mechanisms.


European Journal of Pharmacology | 1996

Melatonin analogues as agonists and antagonists in the circadian system and other brain areas

Shui-Wang Ying; Benjamin Rusak; Philippe Delagrange; Elisabeth Mocaër; Pierre Renard; Beatrice Guardiola-Lemaitre

We studied the effects of drugs related to melatonin on neuronal firing activity in the suprachiasmatic nucleus, intergeniculate leaflet and other brain areas in urethane-anesthetized Syrian hamsters. We tested melatonin and two naphthalenic derivatives of melatonin, a putative agonist (S20098: N-[2-(7-methoxy-1-naphthyl)ethyl]acetamide), and a putative antagonist (S20928: N-[2-(1-naphthyl)ethyl]cyclobutyl carboxamide). Both melatonin and S20098 given intraperitoneally (i.p.) were able to suppress firing rates of cells in a similar dose-dependent manner, but the effects of S20098 were longer lasting. Iontophoresis of melatonin dose dependently depressed spontaneous and light-evoked activity of cells in the suprachiasmatic nucleus and intergeniculate leaflet, while iontophoresis of S20098 was relatively ineffective, probably because it is a poorly charged compound. S20928 (2.0-10 mg/kg, i.p.) alone decreased firing rates of light-sensitive cells by 25-50% for 5-30 min in the suprachiasmatic nucleus and intergeniculate leaflet; however, low doses (< 2.0 mg/kg) of S20928 partially blocked the effects of melatonin agonists on most cells. The non-selective serotonin antagonist metergoline did not block the effects of either melatonin agonist. Both melatonin agonists and antagonists were less effective when applied to cells in the hippocampus and dorsal lateral geniculate nucleus. These results indicate that S20098 is an agonist acting probably on melatonin receptors in the Syrian hamster brain. S20928 may have mixed agonist/antagonist properties, but at low doses appears to function as an antagonist at melatonin receptors in the suprachiasmatic nucleus and intergeniculate leaflet.


The International Journal of Neuropsychopharmacology | 2012

Beneficial behavioural and neurogenic effects of agomelatine in a model of depression/anxiety

Quentin Rainer; Lin Xia; Jean-Philippe Guilloux; Cecilia Gabriel; Elisabeth Mocaër; René Hen; Erika Enhamre; Alain M. Gardier; Denis J. David

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.


Behavioural Brain Research | 2011

Agomelatine reverses the decrease in hippocampal cell survival induced by chronic mild stress.

Girstautė Dagytė; Ilaria Crescente; Folkert Postema; Laure Seguin; Cecilia Gabriel; Elisabeth Mocaër; Johan A. den Boer; Jaap M. Koolhaas

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.


Neuropharmacology | 2004

Differential effects of the novel antidepressant agomelatine (S 20098) versus fluoxetine on 5-HT1A receptors in the rat brain

Naima Hanoun; Elisabeth Mocaër; Pierre-Alain Boyer; Michel Hamon; Laurence Lanfumey

Agomelatine (S 20098) is a novel antidepressant drug with melatonin receptor agonist and 5-HT(2C) receptor antagonist properties, but actual mechanisms underlying its antidepressant action are unknown. Because functional desensitization of 5-HT(1A) autoreceptors in the dorsal raphe nucleus (DRN) occurs after chronic administration of several classes of antidepressants, we investigated whether this adaptive change could also be induced by agomelatine. Neither acute nor chronic treatment with agomelatine (10 mg/kg i.p. for 14 days or 50 mg/kg i.p. for 21 days) changed the density of 5-HT(1A) receptors and their coupling with G proteins in the DRN and the hippocampus in rats. Moreover, these treatments did not affect the basal electrophysiological characteristics and the responses to 5-HT(1A) receptor stimulation of DRN and hippocampal neurons in brain slices. Parallel experiments with melatonin (10 mg/kg i.p. for 14 days) and fluoxetine (5 mg/kg i.p. for 14 days) as reference compounds showed that the former was unable to affect 5-HT(1A) receptors whereas the latter decreased both the 5-HT(1A) receptor-mediated [(35)S]GTP-gamma-S binding and the potency of ipsapirone, a 5-HT(1A) receptor agonist, to inhibit neuronal firing in the DRN. These data indicate that the antidepressant action of agomelatine is not mediated through the same mechanisms as SSRIs or tricyclics.


Neuropsychopharmacology | 2000

Somatodendritic 5-HT1A autoreceptors mediate the anti-aggressive actions of 5-HT1A receptor agonists in rats: An ethopharmacological study with S-15535, alnespirone, and WAY-100635

Sietse F. de Boer; Monique Lesourd; Elisabeth Mocaër; Jaap M. Koolhaas

To elucidate the relative contribution of somatodendritic 5-HT1A autoreceptors and postsynaptic 5-HT1A receptors in the specific anti-aggressive properties of 5-HT1A receptor agonists, the influence of the novel benzodioxopiperazine compound S-15535, which behaves in vivo as a competitive antagonist at postsynaptic 5-HT1A receptors and as an agonist at 5-HT1A autoreceptors, upon offensive and defensive aggression was investigated in wild-type rats using a resident-intruder paradigm. S-15535 exerted a potent dose-dependent decrease in offensive, but not defensive, aggressive behavior (inhibitory dose (ID)50 = 1.11 mg/kg). This anti-aggressive profile was roughly similar to that of the potent pre- and postsynaptic 5-HT1A full agonist alnespirone (ID50 = 1.24). The drugs profound anti-aggressive actions were not accompanied by sedative side effects or signs of the “5-HT1A receptor-mediated behavioral syndrome,” which are characteristically induced by prototypical 5-HT1A receptor agonists like 8-OH-DPAT and buspirone. The selective pre- and postsynaptic 5-HT1A antagonist WAY-100635, which was inactive given alone, abolished the anti-aggressive effects of S-15535 and alnespirone, thereby confirming the involvement of 5-HT1A receptors. Furthermore, combined administration of S-15535 and alnespirone elicited an additive anti-aggressive effect, providing further support for somatodendritic 5-HT1A receptor involvement. Finally, the postsynaptic 5-HT1A antagonistic properties of S-15535 were confirmed by showing blockade of the alnespirone-induced hypothermia, a postsynaptic 5-HT1A mediated response in the rat. These data provide extensive evidence that the anti-aggressive effects of 5-HT1A receptor agonists are expressed via their action on somatodendritic 5-HT1A autoreceptors, thereby most likely attenuating intruder-activated serotonergic neurotransmission.

Collaboration


Dive into the Elisabeth Mocaër's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge