Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Encephale ; 48(2): 179-187, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34649711

ABSTRACT

Recent contradictory data has renewed discussion regarding the existence of adult hippocampal neurogenesis (AHN) in humans, i.e., the continued production of new neurons in the brain after birth. The present review revisits the debate of AHN in humans from a historical point of view in the face of contradictory evidence, analyzing the methods employed to investigate this phenomenon. Thus, to date, of the 57 studies performed in humans that we reviewed, 84% (48) concluded in favor of the presence of newborn neurons in the human adult hippocampus. Besides quality of the tissue (such as postmortem intervals below 26hours as well as tissue conservation and fixation), considerations for assessing and quantify AHN in the human brain require the use of stereology and toxicological analyses of clinical data of the patient.


Subject(s)
Hippocampus , Neurogenesis , Adult , Hippocampus/physiology , Humans , Infant, Newborn , Neurogenesis/physiology , Neurons/physiology
2.
Encephale ; 44(3): 264-273, 2018 Jun.
Article in French | MEDLINE | ID: mdl-29801770

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed antidepressant treatment for treat major depressive disorders. Despite their effectiveness, only 30% of SSRI-treated patients reach remission of depressive symptoms. SSRIs by inhibiting the serotonin transporter present some limits with residual symptoms. Increasing not only serotonin but also norepinephrine and dopamine levels in limbic areas seems to improve remission. Anatomical relationships across serotoninergic, dopaminergic and noradrenergic systems suggest tight reciprocal regulations among them. This review attempts to present, from acute to chronic administration the consequences of SSRI administration on monoaminergic neurotransmission. The serotonin neurons located in the raphe nucleus (RN) are connected to the locus coeruleus (locus coeruleus), the key structure of norepinephrine synthesis, through GABAergic-inhibiting interneurons. Activation of the 5-HT2A receptors expressed on GABAergic interneurons following SERT-inhibition induces an increase in serotonin leading to inhibitory effect on NE release. Similarly, the serotonin neurons exert negative regulation on dopaminergic neurons from the ventral tegmental area (VTA) through a GABAergic interneuron. These interneurons express the 5-HT2C and 5-HT3 receptors inducing an inhibitory effect of 5-HT on DA release. Positive reciprocal connections are also observed through direct projections from the locus coeruleus to the RN and from the VTA to the RN through α1 and D2 receptors respectively, both stimulating the serotoninergic activity. Acute SSRI treatment induces only a slight increase in 5-HT levels in limbic areas due to the activation of presynaptic 5-HT1A and 5-HT1B autoreceptors counteracting the effects of the transporter blockade. No change in NE levels and a small decrease in the dopaminergic neurotransmission is also observed. These weak changes in monoamine in the limbic areas after acute SSRI treatment seems to be one of key point involved in the onset of action. Following desensitization of the 5-HT1A and 5-HT1B autoreceptors, chronic SSRI treatment induces a large increase in the 5-HT neurotransmission. Changes in 5-HT levels at the limbic areas results in a decrease in NE transmission and an increase in DA transmission through an increase in the post-synaptic D2 receptors sensitivity and not from a change in DA levels, which is mainly due to a desensitization of the 5-HT2A receptor. The observed decrease of NE neurotransmission could explain some limits of the SSRI therapy and the interest to activate NE system for producing more robust effects. On the other hand, the D2 sensitization, especially in the nucleus accumbens, stimulates the motivation behavior as well as remission of anhedonia considering the major role of DA release in this structure. Finally, we need to take into account the key role of each monoaminergic neurotransmission to reach remission. Targeting only one system will limit the therapeutic effectiveness. Clinical evidences, including the STAR*D studies, confirmed this by an increase of the remission rate following the mobilization of several monoaminergic transmissions. However, these combinations cannot constitute first line of treatment considering the observed increase of side effects. Such an approach should be adapted to each patient in regard to its particular symptoms as well as clinical history. The next generation of antidepressant therapy will need to take into consideration the interconnections and the interrelation between the monoaminergic systems.


Subject(s)
Antidepressive Agents/pharmacology , Biogenic Monoamines/physiology , Receptor Cross-Talk/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/therapeutic use , Animals , Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Humans , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use
3.
Encephale ; 42(2): 156-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26995153

ABSTRACT

BACKGROUND: Since the discovery of antidepressants, new treatments have emerged with fewer side effects but no greater efficacy. Glycogen synthase kinase 3 ß (GSK-3ß), a kinase known for its activity on glycogen synthesis, has in the last few years raised growing interest in biological psychiatry. Several efficient treatments in major depression have an inhibitory effect on this kinase, which could be targeted in new mood disorder treatments. METHODS: The aim of this review is to summarize findings concerning the intracellular pharmacologic effects of GSK-3ß inhibitors on mood. After a brief description of the intracellular transduction pathways implicated in both GSK-3ß and mood disorders, we reviewed the results demonstrating GSK-3ß involvement in the effects of lithium and ketamine. RESULTS: GSK-3ß can be inhibited through several mechanisms such as serine phosphorylation or binding in a proteic scaffold and others. Its inhibition is implicated in numerous cellular pathways of interest involved in neuronal growth and architecture, cell survival, neurogenesis or synaptic plasticity. This inhibition appears to be both efficient and sufficient in improving mood in animal models. In human beings, several levels of evidence show GSK-3ß inhibition with antidepressant use. Crucially, strong inhibition has been shown with lithium via the proteic scaffold PP2A/ß-arrestin/AKT, and with the rapid antidepressant effect of ketamine via p70S6K. CONCLUSION: Our review focuses on mechanisms whereby the GSK-3ß pathway has a part in the antidepressant effect of lithium and ketamine. This article highlights the importance of translational research from cell and animal models to the clinical setting in order to develop innovative therapeutic targets.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/drug therapy , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Ketamine/pharmacology , Lithium/pharmacology , Antidepressive Agents/pharmacology , Depressive Disorder, Treatment-Resistant/enzymology , Enzyme Inhibitors/pharmacology , Humans , Mood Disorders/drug therapy , Mood Disorders/enzymology
4.
Encephale ; 42(1 Suppl 1): 1S12-23, 2016 Feb.
Article in French | MEDLINE | ID: mdl-26879252

ABSTRACT

Selective Serotonin Reuptake Inhibitors (SSRIs) are extensively used for the treatment of major depressive disorder (MDD). SSRIs are defined as indirect receptor agonists since the activation of postsynaptic receptors is a consequence of an increase in extracellular concentrations of serotonin (5-HT) mediated by the blockade of serotonin transporter. The activation of some serotoninergic receptors (5-HT1A, post-synaptic, 5-HT1B post-synaptic, 5-HT2B, and 5-HT4), but not all (5-HT1A, pre-synaptic, 5-HT1B pre-synaptic, 5-HT2A, 5-HT2C, 5-HT3, and probably 5-HT6), induces anxiolytic/antidepressive - like effects. Targetting specifically some of them could potentially improve the onset of action and/or efficacy and/or prevent MD relapse. Vortioxetine (Brintellix, 1- [2-(2,4-dimethylphenyl-sulfanyl)-phenyl]-piperazine) is a novel multi-target antidepressant drug approved by the Food and Drug Administration (FDA) and by European Medicines Agency. Its properties are markedly different from the extensively prescribed SSRIs. Compared to the SSRIs, vortioxetine is defined as a multimodal antidepressant drug since it is not only a serotonin reuptake inhibitor, but also a 5-HT1D, 5-HT3, 5-HT7 receptor antagonist, 5-HT1B receptor partial agonist and 5-HT1A receptor agonist. This specific pharmacological profile enables vortioxetine to affect not only the serotoninergic and noradrenergic systems, but also the histaminergic, cholinergic, gamma-butyric acid (GABA) ergic and glutamatergic ones. Thus, vortioxetine not only induces antidepressant-like or anxiolytic-like activity but also improves cognitive parameters in several animal models. Indeed, vortioxetine was shown to improve working memory, episodic memory, cognitive flexibility and spatial memory in young adult rodents and also in old animal models. These specific effects of the vortioxetine are of interest considering that cognitive dysfunction is a common comorbidity to MDD. Altogether, even though this molecule still needs to be investigated further, especially in the insufficient-response to antidepressant drugs, vortioxetine is already an innovative therapeutic option for the treatment of major depression.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Piperazines/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sulfides/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Cognition/drug effects , Depressive Disorder, Major/drug therapy , Humans , Receptors, Serotonin/drug effects , Vortioxetine
5.
Neuroscience ; 274: 357-68, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24909899

ABSTRACT

Sleep/wake disorders are frequently associated with anxiety and depression and to elevated levels of cortisol. Even though these alterations are increasingly sought in animal models, no study has investigated the specific effects of chronic corticosterone (CORT) administration on sleep. We characterized sleep/wake disorders in a neuroendocrine mouse model of anxiety/depression, based on chronic CORT administration in the drinking water (35 µg/ml for 4 weeks, "CORT model"). The CORT model was markedly affected during the dark phase by non-rapid eye movement sleep (NREM) increase without consistent alteration of rapid eye movement (REM) sleep. Total sleep duration (SD) and sleep efficiency (SE) increased concomitantly during both the 24h and the dark phase, due to the increase in the number of NREM sleep episodes without a change in their mean duration. Conversely, the total duration of wake decreased due to a decrease in the mean duration of wake episodes despite an increase in their number. These results reflect hypersomnia by intrusion of NREM sleep during the active period as well as a decrease in sleep/wake continuity. In addition, NREM sleep was lighter, with an increased electroencephalogram (EEG) theta activity. With regard to REM sleep, the number and the duration of episodes decreased, specifically during the first part of the light period. REM and NREM sleep changes correlated respectively with the anxiety and the anxiety/depressive-like phenotypes, supporting the notion that studying sleep could be of predictive value for altered emotional behavior. The chronic CORT model in mice that displays hallmark characteristics of anxiety and depression provides an insight into understanding the changes in overall sleep architecture that occur under pathological conditions.


Subject(s)
Anxiety Disorders/physiopathology , Depressive Disorder/physiopathology , Disease Models, Animal , Disorders of Excessive Somnolence/physiopathology , Animals , Brain/physiopathology , Corticosterone , Darkness , Electroencephalography , Emotions , Male , Mice, Inbred C57BL , Photoperiod , Sleep, REM/physiology , Theta Rhythm , Wakefulness
6.
Mol Psychiatry ; 17(11): 1130-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-21912391

ABSTRACT

Women are twice as likely as men to develop major depressive disorder (MDD) and are more prone to recurring episodes. Hence, we tested the hypothesis that the illness may associate with robust molecular changes in female subjects, and investigated large-scale gene expression in the post-mortem brain of MDD subjects paired with matched controls (n=21 pairs). We focused on the lateral/basolateral/basomedian complex of the amygdala as a neural hub of mood regulation affected in MDD. Among the most robust findings were downregulated transcripts for genes coding for γ-aminobutyric acid (GABA) interneuron-related peptides, including somatostatin (SST), tachykinin, neuropeptide Y (NPY) and cortistatin, in a pattern reminiscent to that previously reported in mice with low brain-derived neurotrophic factor (BDNF). Changes were confirmed by quantitative PCR and not explained by demographic, technical or known clinical parameters. BDNF itself was significantly downregulated at the RNA and protein levels in MDD subjects. Investigating putative mechanisms, we show that this core MDD-related gene profile (including SST, NPY, TAC1, RGS4 and CORT) is recapitulated by complementary patterns in mice with constitutive (BDNF-heterozygous) or activity-dependent (exon IV knockout) decreases in BDNF function, with a common effect on SST and NPY. Together, these results provide both direct (low RNA/protein) and indirect (low BDNF-dependent gene pattern) evidence for reduced BDNF function in the amygdala of female subjects with MDD. Supporting studies in mutant mice models suggest a complex mechanism of low constitutive and activity-dependent BDNF function in MDD, particularly affecting SST/NPY-related GABA neurons, thus linking the neurotrophic and GABA hypotheses of depression.


Subject(s)
Amygdala/metabolism , Brain-Derived Neurotrophic Factor/genetics , Depressive Disorder, Major/genetics , GABAergic Neurons/metabolism , Adolescent , Adult , Aged , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Case-Control Studies , Down-Regulation/genetics , Female , Gene Expression Profiling/methods , Genetic Association Studies/methods , Humans , Mice , Mice, Knockout , Middle Aged , Neuropeptide Y/genetics , Neuropeptides/genetics , Somatostatin/genetics , Tachykinins/genetics
8.
Neuropharmacology ; 55(6): 1006-14, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18761360

ABSTRACT

Antidepressants such as Selective Serotonin Reuptake Inhibitors (SSRI) act as indirect agonists of serotonin (5-HT) receptors. Although these drugs produce a rapid blockade of serotonin transporters (SERTs) in vitro, several weeks of treatment are necessary to observe clinical benefits. This paradox has not been solved yet. Recent studies have identified modifications of intracellular signaling proteins and target genes that could contribute to antidepressant-like activity of SSRI (e.g., increases in neurogenesis and BDNF protein levels), and may explain, at least in part, their long delay of action. Although these data suggest a positive regulation of 5-HT on the expression of the gene coding for BDNF, the reciprocal effects of BDNF on brain 5-HT neurotransmission remains poorly documented. To study the impact of BDNF on serotonergic activity, a dual experimental strategy was used to analyze neurochemical and behavioral consequences of its decrease (strategy 1) or increase (strategy 2) in the brain of adult male mice. (1) In heterozygous BDNF+/- mice in which brain BDNF protein levels were decreased by half, an enhancement of basal extracellular 5-HT levels (5-HText) that induced a down-regulation of SERT, i.e., a decrease in its capacity to reuptake 5-HT, was found in the hippocampus. In addition, the SSRI, paroxetine, failed to increase hippocampal 5-HText in BDNF+/- mice, while it produces robust effects in wild-type littermates. Thus, BDNF+/- mice can be viewed as an animal model of genetic resistance to serotonergic antidepressant drugs. (2) In wild-type BDNF+/+ mice, the effects of intra-hippocampal (vHi) injection of BDNF (100 ng) in combination with a SSRI was examined by using intracerebral microdialysis and behavioral paradigms that predict an antidepressant- and anxiolytic-like activity of a molecule [the forced swim test (FST) and the open field paradigm (OF) respectively]. BDNF induced a rapid and transient increase in paroxetine response on 5-HText in the adult hippocampus, which was correlated with a potentiation of its antidepressant-like activity in the FST. The effects of BDNF were selectively blocked by K252a, an antagonist of its high-affinity TrkB receptor. Such a correlation between neurochemical and behavioral effects of [BDNF+SSRI] co-administration suggests that its antidepressant-like activity is linked to the activation of 5-HT neurotransmission in the adult hippocampus. BDNF also had a facilitatory effect on anxiety-like behavior in the OF test, and paroxetine prevented this anxiogenesis. What was the mechanism by which BDNF exerted these latter effects? Surprisingly, by using zero net flux method of quantitative microdialysis in vivo, we found that an intra-hippocampal BDNF injection in wild-type mice decreased the functional activity of SERT as observed in BDNF+/- mice. However, the decreased capacity of SERT to reuptake 5-HT was not associated to an increase in basal 5-HText in the hippocampus of WT mice. Interestingly, using in situ hybridization experiments indicated that TrkB receptor mRNA was expressed in the hippocampus and dorsal raphe nucleus in adult mice suggesting that the neurochemical and behavioral effects of intra-hippocampal BDNF injection can mobilize both pre- and post-synaptic elements of the brain 5-HT neurotransmission. Taken together, these set of experiments unveiled a relative opposition of neurochemical and behavioral responses following either a decrease (in BDNF+/- mutant mice) or an increase in brain BDNF levels (bilateral intra-hippocampal injection) in adult mice. In view of developing new antidepressant drug strategy, a poly-therapy combining BDNF with a chronic SSRI treatment could thus improve the efficacy of current medications.


Subject(s)
Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Gene Expression Regulation/physiology , Hippocampus/metabolism , Serotonin/metabolism , Animals , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Brain-Derived Neurotrophic Factor/deficiency , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Mice , Mice, Knockout , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
9.
J Mol Neurosci ; 30(1-2): 105-6, 2006.
Article in English | MEDLINE | ID: mdl-17192649

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) in the brain exhibit diverse functional properties and ubiquitous distribution. Yet, except for providing a receptor for the exogenously applied nicotine of tobacco products, their role in the normal functioning of the brain has remained elusive. We have used a lentiviral expression vector to re-express the beta2 subunit specifically in the ventral tegmental area (VTA) of beta2-/- mice. The viral vector efficiently expresses beta2- subunit protein leading to new nAChR-binding sites. VTA neurons transduced by the lentiviral vector are responsive to intravenous nicotine when analyzed using in vivo electrophysiology. Nicotine-induced dopamine release from the nucleus accumbens (NuAcc) was also restored in re-expressing beta2-/- mice. Intra-VTA injection of nicotine was found to be reinforcing in both wild-type and beta2-subunit re-expressing beta2-/- mice, but not in beta2-/- mice. Furthermore, in the absence of applied nicotine, the spontaneous slow exploratory behavior of the mice was restored, whereas fast navigation did not change. This latter behavioral analysis suggests a role for beta2* nAChR, specifically expressed in the VTA, in mammalian cognitive function.


Subject(s)
Brain/physiology , Genetic Vectors , Lentivirus/genetics , Receptors, Nicotinic/genetics , Animals , Behavior, Addictive/genetics , Cognition/physiology , Exploratory Behavior , Mice , Mice, Knockout , Nicotine , Receptors, Nicotinic/deficiency , Recombinant Proteins/metabolism
10.
Nature ; 436(7047): 103-7, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-16001069

ABSTRACT

Worldwide, 100 million people are expected to die this century from the consequences of nicotine addiction, but nicotine is also known to enhance cognitive performance. Identifying the molecular mechanisms involved in nicotine reinforcement and cognition is a priority and requires the development of new in vivo experimental paradigms. The ventral tegmental area (VTA) of the midbrain is thought to mediate the reinforcement properties of many drugs of abuse. Here we specifically re-expressed the beta2-subunit of the nicotinic acetylcholine receptor (nAChR) by stereotaxically injecting a lentiviral vector into the VTA of mice carrying beta2-subunit deletions. We demonstrate the efficient re-expression of electrophysiologically responsive, ligand-binding nicotinic acetylcholine receptors in dopamine-containing neurons of the VTA, together with the recovery of nicotine-elicited dopamine release and nicotine self-administration. We also quantified exploratory behaviours of the mice, and showed that beta2-subunit re-expression restored slow exploratory behaviour (a measure of cognitive function) to wild-type levels, but did not affect fast navigation behaviour. We thus demonstrate the sufficient role of the VTA in both nicotine reinforcement and endogenous cholinergic regulation of cognitive functions.


Subject(s)
Cognition/physiology , Gene Expression , Nicotine/metabolism , Receptors, Nicotinic/metabolism , Animals , Cognition/drug effects , Dopamine/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Locomotion/physiology , Mice , Morphine/administration & dosage , Morphine/pharmacology , Neurons/drug effects , Neurons/metabolism , Nicotine/administration & dosage , Nicotine/pharmacology , Receptors, Adrenergic, beta-2/deficiency , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
11.
J Neurosci Methods ; 140(1-2): 53-7, 2004 Dec 30.
Article in English | MEDLINE | ID: mdl-15589334

ABSTRACT

An important concern about microdialysis methodology is the histological validation of the dialysis probe implantation site in brain tissue of rodents (rat, mouse). Several methods have been described on standard histological staining (i.e., cresyl violet, formalin fixation, fast green perfusion, etc.). However, this methodology is time consuming. These requirements are not compatible with a histological validation prior to analysis of microdialysis samples. Here, we developed a new method to locate the track of the dialysis probe in the rodent brain. This method is based on a digital photomicrograph of a coronal section of the rodent frozen brain. The fitting of an appropriate coronal diagram of the rats' and mice' brain atlas with this photomicrograph, allowed us to locate precisely and quickly the track of the dialysis probe.


Subject(s)
Anatomy, Artistic/methods , Brain Mapping/methods , Brain/anatomy & histology , Brain/surgery , Medical Illustration , Microdialysis/instrumentation , Microdialysis/methods , Photomicrography/methods , Anatomy, Artistic/instrumentation , Animals , Brain/physiology , Brain Mapping/instrumentation , Male , Mice , Mice, Inbred C57BL , Microelectrodes/standards , Photomicrography/instrumentation , Rats , Rats, Wistar , Species Specificity , Stereotaxic Techniques/trends , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...