Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell ; 149(5): 1152-63, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632977

ABSTRACT

Our understanding of current treatments for depression, and the development of more specific therapies, is limited by the complexity of the circuits controlling mood and the distributed actions of antidepressants. Although the therapeutic efficacy of serotonin-specific reuptake inhibitors (SSRIs) is correlated with increases in cortical activity, the cell types crucial for their action remain unknown. Here we employ bacTRAP translational profiling to show that layer 5 corticostriatal pyramidal cells expressing p11 (S100a10) are strongly and specifically responsive to chronic antidepressant treatment. This response requires p11 and includes the specific induction of Htr4 expression. Cortex-specific deletion of p11 abolishes behavioral responses to SSRIs, but does not lead to increased depression-like behaviors. Our data identify corticostriatal projection neurons as critical for the response to antidepressants, and suggest that the regulation of serotonergic tone in this single cell type plays a pivotal role in antidepressant therapy.


Subject(s)
Antidepressive Agents/metabolism , Depression/drug therapy , Neurons/cytology , Prefrontal Cortex/cytology , Selective Serotonin Reuptake Inhibitors/metabolism , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
2.
Mol Psychiatry ; 26(6): 2334-2349, 2021 06.
Article in English | MEDLINE | ID: mdl-33441982

ABSTRACT

Serotonin receptor 4 (5-HT4R) plays an important role in regulating mood, anxiety, and cognition, and drugs that activate this receptor have fast-acting antidepressant (AD)-like effects in preclinical models. However, 5-HT4R is widely expressed throughout the central nervous system (CNS) and periphery, making it difficult to pinpoint the cell types and circuits underlying its effects. Therefore, we generated a Cre-dependent 5-HT4R knockout mouse line to dissect the function of 5-HT4R in specific brain regions and cell types. We show that the loss of functional 5-HT4R specifically from excitatory neurons of hippocampus led to robust AD-like behavioral responses and an elevation in baseline anxiety. 5-HT4R was necessary to maintain the proper excitability of dentate gyrus (DG) granule cells and cell type-specific molecular profiling revealed a dysregulation of genes necessary for normal neural function and plasticity in cells lacking 5-HT4R. These adaptations were accompanied by an increase in the number of immature neurons in ventral, but not dorsal, dentate gyrus, indicating a broad impact of 5-HT4R loss on the local cellular environment. This study is the first to use conditional genetic targeting to demonstrate a direct role for hippocampal 5-HT4R signaling in modulating mood and anxiety. Our findings also underscore the need for cell type-based approaches to elucidate the complex action of neuromodulatory systems on distinct neural circuits.


Subject(s)
Anxiety , Hippocampus , Animals , Dentate Gyrus/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Receptors, Serotonin , Receptors, Serotonin, 5-HT4/genetics , Receptors, Serotonin, 5-HT4/metabolism
3.
Hum Mol Genet ; 26(1): 192-209, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28082376

ABSTRACT

Local mRNA translation in growing axons allows for rapid and precise regulation of protein expression in response to extrinsic stimuli. However, the role of local translation in mature CNS axons is unknown. Such a mechanism requires the presence of translational machinery and associated mRNAs in circuit-integrated brain axons. Here we use a combination of genetic, quantitative imaging and super-resolution microscopy approaches to show that mature axons in the mammalian brain contain ribosomes, the translational regulator FMRP and a subset of FMRP mRNA targets. This axonal translational machinery is associated with Fragile X granules (FXGs), which are restricted to axons in a stereotyped subset of brain circuits. FXGs and associated axonal translational machinery are present in hippocampus in humans as old as 57 years. This FXG-associated axonal translational machinery is present in adult rats, even when adult neurogenesis is blocked. In contrast, in mouse this machinery is only observed in juvenile hippocampal axons. This differential developmental expression was specific to the hippocampus, as both mice and rats exhibit FXGs in mature axons in the adult olfactory system. Experiments in Fmr1 null mice show that FMRP regulates axonal protein expression but is not required for axonal transport of ribosomes or its target mRNAs. Axonal translational machinery is thus a feature of adult CNS neurons. Regulation of this machinery by FMRP could support complex behaviours in humans throughout life.


Subject(s)
Axons/pathology , Brain/pathology , Cytoplasmic Granules/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/pathology , RNA, Messenger/metabolism , Ribosomes/pathology , Adult , Animals , Axons/metabolism , Brain/metabolism , Cytoplasmic Granules/pathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurogenesis/genetics , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Ribosomes/metabolism
4.
Proc Natl Acad Sci U S A ; 109(28): 11360-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733786

ABSTRACT

A large number of studies have demonstrated that the nucleus accumbens (NAC) is a critical site in the neuronal circuits controlling reward responses, motivation, and mood, but the neuronal cell type(s) underlying these processes are not yet known. Identification of the neuronal cell types that regulate depression-like states will guide us in understanding the biological basis of mood and its regulation by diseases like major depressive disorder. Taking advantage of recent findings demonstrating that the serotonin receptor chaperone, p11, is an important molecular regulator of depression-like states, here we identify cholinergic interneurons (CINs) as a primary site of action for p11 in the NAC. Depression-like behavior is observed in mice after decrease of p11 levels in NAC CINs. This phenotype is recapitulated by silencing neuronal transmission in these cells, demonstrating that accumbal cholinergic neuronal activity regulates depression-like behaviors and suggesting that accumbal CIN activity is crucial for the regulation of mood and motivation.


Subject(s)
Annexin A2/metabolism , Depression/physiopathology , Interneurons/metabolism , Nucleus Accumbens/metabolism , S100 Proteins/metabolism , Acetylcholine/metabolism , Animals , Antidepressive Agents/pharmacology , Behavior, Animal , Depression/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Chaperones/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Phenotype , Receptors, Cholinergic/metabolism
5.
Proc Natl Acad Sci U S A ; 108(22): 9262-7, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21518864

ABSTRACT

Antiinflammatory drugs achieve their therapeutic actions at least in part by regulation of cytokine formation. A "cytokine hypothesis" of depression is supported by the observation that depressed individuals have elevated plasma levels of certain cytokines compared with healthy controls. Here we investigated a possible interaction between antidepressant agents and antiinflammatory agents on antidepressant-induced behaviors and on p11, a biochemical marker of depressive-like states and antidepressant responses. We found that widely used antiinflammatory drugs antagonize both biochemical and behavioral responses to selective serotonin reuptake inhibitors (SSRIs). In contrast to the levels detected in serum, we found that frontal cortical levels of certain cytokines (e.g., TNFα and IFNγ) were increased by serotonergic antidepressants and that these effects were inhibited by antiinflammatory agents. The antagonistic effect of antiinflammatory agents on antidepressant-induced behaviors was confirmed by analysis of a dataset from a large-scale real-world human study, "sequenced treatment alternatives to relieve depression" (STAR*D), underscoring the clinical significance of our findings. Our data indicate that clinicians should carefully balance the therapeutic benefits of antiinflammatory agents versus the potentially negative consequences of antagonizing the therapeutic efficacy of antidepressant agents in patients suffering from depression.


Subject(s)
Annexin A2/metabolism , Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Drug Interactions , S100 Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Citalopram/pharmacology , Cytokines/metabolism , Fluoxetine/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL
6.
Proc Natl Acad Sci U S A ; 107(9): 4401-6, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20145109

ABSTRACT

Dopamine neurotransmission controls motor and perseverative behavior, is mediated by protein phosphorylation, and may be perturbed in disorders of attention and hyperactivity. To assess the role of casein kinase I (CK1) in the regulation of dopamine signaling, we generated a genetically modified mouse line that overexpresses CK1delta (CK1delta OE) specifically in the forebrain. Overexpression was confirmed both at the mRNA and at the protein levels. Under basal conditions, CK1delta OE mice exhibited horizontal and vertical hyperactivity, reduced anxiety, and nesting behavior deficiencies. The CK1delta OE mice also presented paradoxical responses to dopamine receptor stimulation, showing hypoactivity following injection of d-amphetamine or methylphenidate, indicating that CK1 activity has a profound effect on dopamine signaling in vivo. Interestingly, CK1delta overexpression led to significantly reduced D1R and D2R dopamine receptor levels. All together, under basal conditions and in response to drug stimulation, the behavioral phenotype of CK1delta OE mice is reminiscent of the symptoms and drug responses observed in attention-deficit/hyperactivity disorder and therefore the CK1delta OE mice appear to be a model for this disorder.


Subject(s)
Attention Deficit Disorder with Hyperactivity/metabolism , Casein Kinase Idelta/metabolism , Down-Regulation , Locomotion , Prosencephalon/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Amphetamine/pharmacology , Animals , Attention Deficit Disorder with Hyperactivity/enzymology , Behavior, Animal/drug effects , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Methylphenidate/pharmacology , Mice
7.
Proc Natl Acad Sci U S A ; 105(32): 11352-7, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18682560

ABSTRACT

All classes of antidepressants increase hippocampal cell proliferation and neurogenesis, which contributes, in part, to the behavioral actions of these treatments. Among antidepressant treatments, electroconvulsive seizure (ECS) is the most robust stimulator of hippocampal cell proliferation and the most efficacious treatment for depression, but the cellular mechanisms underlying the actions of ECS are unknown. To address this question, we investigated the effect of ECS on proliferation of neural stem-like and/or progenitor cells in the subgranular zone of rat dentate gyrus. We define the neural differentiation cascade from stem-like cells to early neural progenitors (also referred to as quiescent and amplifying neural progenitors, respectively) by coexpression of selective cellular and mitotic activity markers. We find that at an early mitotic phase ECS increases the proliferation of quiescent progenitors and then at a later phase increases the proliferation of amplifying progenitors. We further demonstrate that vascular endothelial growth factor (VEGF) signaling is necessary for ECS induction of quiescent neural progenitor cell proliferation and is sufficient to produce this effect. These findings demonstrate that ECS and subsequent induction of VEGF stimulates the proliferation of neural stem-like cells and neural progenitor cells, thereby accounting for the superior neurogenic actions of ECS compared with chemical antidepressants.


Subject(s)
Cell Proliferation , Dentate Gyrus/metabolism , Electroshock , Neurons/metabolism , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Antigens, Differentiation/biosynthesis , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/pathology , Depression/therapy , Gene Expression Regulation/drug effects , Male , Neurons/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stem Cells/pathology , Vascular Endothelial Growth Factor A/biosynthesis
8.
J Neurosci ; 29(6): 1937-46, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19211900

ABSTRACT

p11 (S100A10), a member of a large family of S100 proteins, interacts with serotonin receptor 1B (5-HTR1B), modulates 5-HT1B receptor signal transduction, and is required for antidepressant responses to activation of this receptor. In the current study, we investigated the specificity of the interaction between 5-HTR1B and p11 by screening brain-expressed S100 proteins against serotonin and noradrenergic receptors. The data indicate that p11 is unique among its family members for its interactions with defined serotonin receptors. We identify a novel p11-interacting receptor (5-HTR4) and characterize the interaction between p11 and 5-HTR4, demonstrating that (1) p11 and 5-HTR4 mRNA and protein are coexpressed in brain regions that are relevant for major depression, (2) p11 increases 5-HTR4 surface expression and facilitates 5-HTR4 signaling, and (3) p11 is required for the behavioral antidepressant responses to 5-HTR4 stimulation in vivo. The essential role played by p11 in modulating signaling through 5-HT4 as well as 5-HT1B receptors supports the concept that this protein may be a key determinant of vulnerability to depression.


Subject(s)
Annexin A2/physiology , Apoptosis/physiology , Cell Membrane/physiology , Exploratory Behavior/physiology , Receptors, Serotonin, 5-HT4/physiology , Receptors, Serotonin/physiology , S100 Proteins/physiology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Depressive Disorder/metabolism , Mice , Mice, Inbred C57BL
9.
Curr Opin Pharmacol ; 8(1): 14-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18061540

ABSTRACT

Antidepressants are among the most widely prescribed drugs, however the mechanism underlying their therapeutic efficacy is not known. Neurotrophic factors represent a promising class of targets for antidepressant treatments. We recently characterized a role for vascular endothelial growth factor (VEGF) in cellular and behavioral antidepressant responses. VEGF is a potent mitogen and survival factor for endothelial cells (ECs) and neurons, and modulator of synaptic transmission. Because VEGF has been implicated in a variety of diseases, understanding the molecular and cellular specificity of antidepressant-induced VEGF will be crucial to determine its potential as a therapeutic target in depression.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Vascular Endothelial Growth Factor A/physiology , Humans , Neovascularization, Physiologic , Neurons/physiology
10.
Front Behav Neurosci ; 13: 141, 2019.
Article in English | MEDLINE | ID: mdl-31293404

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and autism. FXS is also accompanied by attention problems, hyperactivity, anxiety, aggression, poor sleep, repetitive behaviors, and self-injury. Recent work supports the role of γ-aminobutyric-acid (GABA), the primary inhibitory neurotransmitter in the brain, in mediating symptoms of FXS. Deficits in GABA machinery have been observed in a mouse model of FXS, including a loss of tonic inhibition in the amygdala, which is mediated by extrasynaptic GABAA receptors. Humans with FXS also show reduced GABAA receptor availability. Here, we sought to evaluate the potential of gaboxadol (also called OV101 and THIP), a selective and potent agonist for delta-subunit-containing extrasynaptic GABAA receptors (dSEGA), as a therapeutic agent for FXS by assessing its ability to normalize aberrant behaviors in a relatively uncharacterized mouse model of FXS (Fmr1 KO2 mice). Four behavioral domains (hyperactivity, anxiety, aggression, and repetitive behaviors) were probed using a battery of behavioral assays. The results showed that Fmr1 KO2 mice were hyperactive, had abnormal anxiety-like behavior, were more irritable and aggressive, and had an increased frequency of repetitive behaviors compared to wild-type (WT) littermates, which are all behavioral deficits reminiscent of individuals with FXS. Treatment with gaboxadol normalized all of the aberrant behaviors observed in Fmr1 KO2 mice back to WT levels, providing evidence of its potential benefit for treating FXS. We show that the potentiation of extrasynaptic GABA receptors alone, by gaboxadol, is sufficient to normalize numerous behavioral deficits in the FXS model using endpoints that are directly translatable to the clinical presentation of FXS. Taken together, these data support the future evaluation of gaboxadol in individuals with FXS, particularly with regard to symptoms of hyperactivity, anxiety, irritability, aggression, and repetitive behaviors.

11.
Eur J Neurosci ; 27(6): 1485-93, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18336568

ABSTRACT

Ongoing neurogenesis in the adult hippocampus is thought to play a role in learning and memory processes, and in response to antidepressant treatments. Low doses of irradiation (IRR) produce a significant long-lasting inhibitory effect on hippocampal neurogenesis that correlates with long-lasting behavioral deficits. Here we report that electroconvulsive seizure (ECS), which robustly increases adult neurogenesis in naïve animals, also reverses the disruption of neurogenesis produced by IRR exposure. Moreover, we find that vascular endothelial growth factor (VEGF) is an essential mediator of this effect. Expression of VEGF in the granule cell layer (GCL) of the hippocampus is decreased by IRR, and ECS administration reverses this deficit in VEGF. There is a corresponding alteration in the number of endothelial cells, which express VEGF, in the hippocampal GCL following IRR and ECS. We also find that blockade of VEGF signaling attenuates ECS-induced proliferation, and VEGF infusion partially restores proliferation in irradiated animals. To examine the functional consequences of IRR and ECS on neurogenesis, hippocampus-dependent contextual fear conditioning was assessed. We found that following disruption by IRR, ECS restores contextual learning to baseline levels at time points consistent with its effects on neurogenesis. These findings demonstrate that ECS, in part via induction of VEGF, can reverse long-term neurogenesis deficits resulting from IRR, and that these effects have functional consequences on hippocampus-dependent fear memory.


Subject(s)
Cell Differentiation/radiation effects , Electroshock , Fear/radiation effects , Gamma Rays , Hippocampus/cytology , Hippocampus/radiation effects , Memory/radiation effects , Neurons/radiation effects , Animals , Cell Differentiation/physiology , Electroshock/methods , Fear/physiology , Hippocampus/physiology , Male , Memory/physiology , Neurons/cytology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/physiology
12.
Biol Psychiatry ; 68(6): 528-35, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20591415

ABSTRACT

BACKGROUND: The protein p11 (also called S100A10) is downregulated in human and rodent depressive-like states. Considerable experimental evidence also implicates p11 in the mechanism of action of antidepressant drugs and electroconvulsive seizures, in part due to its interaction with specific serotonin receptors. Brain-derived neurotrophic factor (BDNF) has been linked to the therapeutic activity of antidepressants in rodent models and humans. In the current study, we investigated whether BDNF regulates p11 in vitro and in vivo. METHODS: We utilized primary neuronal cultures, in vivo analyses of transgenic mice, and behavioral techniques to assess the effects of BDNF on p11. RESULTS: Results indicate that BDNF stimulates p11 expression through tropomyosin-related kinase B (trkB) receptors and via the mitogen-activated protein kinase signaling pathway. Brain-derived neurotrophic factor-induced changes in p11 in vivo correlate with changes in ligand binding to the 5-hydroxytryptamine receptor 1B, the subcellular localization of which is known to be regulated by p11. Behavioral studies demonstrate that p11 knockout mice are insensitive to the antidepressant actions of BDNF. CONCLUSIONS: Taken together, our data demonstrate that p11 levels are regulated by BDNF in vitro and in vivo and that the antidepressant-like effect of BDNF in two well-established behavioral models requires p11. These data support a role for p11 in the antidepressant activity of neurotrophins.


Subject(s)
Annexin A2/biosynthesis , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/physiology , Citalopram/pharmacology , Gene Expression Regulation/drug effects , S100 Proteins/biosynthesis , Signal Transduction/drug effects , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/administration & dosage , Brain-Derived Neurotrophic Factor/genetics , Cell Culture Techniques , Dose-Response Relationship, Drug , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Gene Expression Regulation/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Serotonin/pharmacology , Signal Transduction/physiology
13.
Biol Psychiatry ; 65(5): 392-400, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19006787

ABSTRACT

BACKGROUND: Although it has been postulated for many years that depression is associated with loss of synapses, primarily in the hippocampus, and that antidepressants facilitate synapse growth, we still lack ultrastructural evidence that changes in depressive behavior are indeed correlated with structural synaptic modifications. METHODS: We analyzed hippocampal spine synapses of male rats (n=127) with electron microscopic stereology in association with performance in the learned helplessness paradigm. RESULTS: Inescapable footshock (IES) caused an acute and persistent loss of spine synapses in each of CA1, CA3, and dentate gyrus, which was associated with a severe escape deficit in learned helplessness. On the other hand, IES elicited no significant synaptic alterations in motor cortex. A single injection of corticosterone reproduced both the hippocampal synaptic changes and the behavioral responses induced by IES. Treatment of IES-exposed animals for 6 days with desipramine reversed both the hippocampal spine synapse loss and the escape deficit in learned helplessness. We noted, however, that desipramine failed to restore the number of CA1 spine synapses to nonstressed levels, which was associated with a minor escape deficit compared with nonstressed control rats. Shorter, 1-day or 3-day desipramine treatments, however, had neither synaptic nor behavioral effects. CONCLUSIONS: These results indicate that changes in depressive behavior are associated with remarkable remodeling of hippocampal spine synapses at the ultrastructural level. Because spine synapse loss contributes to hippocampal dysfunction, this cellular mechanism may be an important component in the neurobiology of stress-related disorders such as depression.


Subject(s)
Depression/pathology , Escape Reaction/drug effects , Helplessness, Learned , Hippocampus/ultrastructure , Synapses/ultrastructure , Animals , Anti-Inflammatory Agents/pharmacology , Antidepressive Agents, Tricyclic/administration & dosage , Biomarkers, Pharmacological/analysis , Corticosterone/blood , Corticosterone/pharmacology , Depression/blood , Depression/drug therapy , Desipramine/administration & dosage , Disease Models, Animal , Hippocampus/drug effects , Male , Motor Cortex/drug effects , Motor Cortex/ultrastructure , Rats , Rats, Sprague-Dawley , Stress, Physiological , Synapses/drug effects
14.
Proc Natl Acad Sci U S A ; 104(11): 4647-52, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360578

ABSTRACT

The neural mechanisms underlying the cellular and behavioral responses to antidepressants are not yet known. Up-regulation of growth factors and adult neurogenesis suggest a role for one or more of these factors in the action of antidepressants. One candidate of interest is vascular endothelial growth factor (VEGF), which was initially characterized for its role in angiogenesis, but also exerts direct mitogenic effects on neural progenitors in vitro. Results of this study demonstrate that VEGF is induced by multiple classes of antidepressants at time points consistent with the induction of cell proliferation and therapeutic action of these treatments. We find that VEGF signaling through the Flk-1 receptor is required for antidepressant-induced cell proliferation. We also show that VEGF-Flk-1 signaling is required and sufficient for behavioral responses in two chronic and two subchronic antidepressant models. Taken together, these studies identify VEGF and VEGF-Flk-1 signaling as mediators of antidepressant actions and potential targets for therapeutic intervention.


Subject(s)
Antidepressive Agents/pharmacology , Neovascularization, Pathologic , Neurons/metabolism , Vascular Endothelial Growth Factor A/physiology , Animals , Behavior, Animal , Brain/metabolism , Cell Proliferation , Electroshock , Male , Mice , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Hippocampus ; 16(3): 239-49, 2006.
Article in English | MEDLINE | ID: mdl-16425236

ABSTRACT

The hippocampus is one of several limbic brain structures implicated in the pathophysiology and treatment of mood disorders. Preclinical and clinical studies demonstrate that stress and depression lead to reductions of the total volume of this structure and atrophy and loss of neurons in the adult hippocampus. One of the cellular mechanisms that could account for alterations of hippocampal structure as well as function is the regulation of adult neurogenesis. Stress exerts a profound effect on neurogenesis, leading to a rapid and prolonged decrease in the rate of cell proliferation in the adult hippocampus. In contrast, chronic antidepressant treatment up-regulates hippocampal neurogenesis, and could thereby block or reverse the atrophy and damage caused by stress. Recent studies also demonstrate that neurogenesis is required for the actions of antidepressants in behavioral models of depression. This review discusses the literature that has lead to a neurogenic hypothesis of depression and antidepressant action, as well as the molecular and cellular mechanisms that underlie the regulation of adult neurogenesis by stress and antidepressant treatment.


Subject(s)
Antidepressive Agents/pharmacology , Cell Proliferation/drug effects , Dentate Gyrus/physiology , Depressive Disorder/complications , Neuronal Plasticity/physiology , Stress, Physiological/physiopathology , Animals , Atrophy/drug therapy , Atrophy/physiopathology , Dentate Gyrus/cytology , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Humans , Neuronal Plasticity/drug effects , Neurons/metabolism , Stem Cells/metabolism , Stress, Physiological/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL