Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Psychiatry ; 28(11): 4632-4641, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37696873

ABSTRACT

Reductions of astroglia expressing glial fibrillary acidic protein (GFAP) are consistently found in the prefrontal cortex (PFC) of patients with depression and in rodent chronic stress models. Here, we examine the consequences of PFC GFAP+ cell depletion and cell activity enhancement on depressive-like behaviors in rodents. Using viral expression of diphtheria toxin receptor in PFC GFAP+ cells, which allows experimental depletion of these cells following diphtheria toxin administration, we demonstrated that PFC GFAP+ cell depletion induced anhedonia-like behavior within 2 days and lasting up to 8 days, but no anxiety-like deficits. Conversely, activating PFC GFAP+ cell activity for 3 weeks using designer receptor exclusively activated by designer drugs (DREADDs) reversed chronic restraint stress-induced anhedonia-like deficits, but not anxiety-like deficits. Our results highlight a critical role of cortical astroglia in the development of anhedonia and further support the idea of targeting astroglia for the treatment of depression.


Subject(s)
Anhedonia , Astrocytes , Animals , Humans , Astrocytes/metabolism , Prefrontal Cortex/metabolism , Depression/metabolism , Stress, Psychological/metabolism , Behavior, Animal
2.
Res Sq ; 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37461693

ABSTRACT

Reductions of astroglia expressing glial fibrillary acidic protein (GFAP) are consistently found in the prefrontal cortex (PFC) of patients with depression and in rodent chronic stress models. Here, we examine the consequences of PFC GFAP+ cell depletion and cell activity enhancement on depressive-like behaviors in rodents. Using viral expression of diphtheria toxin receptor in PFC GFAP+ cells, which allows experimental depletion of these cells following diphtheria toxin administration, we demonstrated that PFC GFAP+ cell depletion induced anhedonia-like behavior within 2 days and lasting up to 8 days, but no anxiety-like deficits. Conversely, activating PFC GFAP+ cell activity for 3 weeks using designer receptor exclusively activated by designer drugs (DREADDs) reversed chronic restraint stress-induced anhedonia-like deficits, but not anxiety-like deficits. Our results highlight a critical role of cortical astroglia in the development of anhedonia and further support the idea of targeting astroglia for the treatment of depression.

3.
Mol Psychiatry ; 25(11): 3109-3111, 2020 Nov.
Article in English | MEDLINE | ID: mdl-30862939

ABSTRACT

A number of collaborators were not acknowledged for their contribution to this published article. The acknowledgements that were missing in this published article can now be found in the associated correction.

4.
Mol Psychiatry ; 23(4): 963-972, 2018 04.
Article in English | MEDLINE | ID: mdl-28461698

ABSTRACT

Psychotic symptoms, defined as the occurrence of delusions or hallucinations, are frequent in Alzheimer disease (AD), affecting ~40 to 60% of individuals with AD (AD with psychosis (AD+P)). In comparison with AD subjects without psychosis, AD+P subjects have more rapid cognitive decline and poor outcomes. Prior studies have estimated the heritability of psychosis in AD at 61%, but the underlying genetic sources of this risk are not known. We evaluated a Discovery Cohort of 2876 AD subjects with (N=1761) or without psychosis (N=1115). All subjects were genotyped using a custom genotyping array designed to evaluate single-nucleotide polymorphisms (SNPs) with evidence of genetic association with AD+P and include SNPs affecting or putatively affecting risk for schizophrenia and AD. Results were replicated in an independent cohort of 2194 AD subjects with (N=734) or without psychosis (N=1460). We found that AD+P is associated with polygenic risk for a set of novel loci and inversely associated with polygenic risk for schizophrenia. Among the biologic pathways identified by the associations of schizophrenia SNPs with AD+P are endosomal trafficking, autophagy and calcium channel signaling. To the best of our knowledge, these findings provide the first clear demonstration that AD+P is associated with common genetic variation. In addition, they provide an unbiased link between polygenic risk for schizophrenia and a lower risk of psychosis in AD. This provides an opportunity to leverage progress made in identifying the biologic effects of schizophrenia alleles to identify novel mechanisms protecting against more rapid cognitive decline and psychosis risk in AD.


Subject(s)
Alzheimer Disease/genetics , Psychotic Disorders/genetics , Schizophrenia/genetics , Aged , Aged, 80 and over , Alleles , Alzheimer Disease/complications , Alzheimer Disease/psychology , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Multifactorial Inheritance , Polymorphism, Single Nucleotide , Psychotic Disorders/complications , Schizophrenia/complications
6.
Mol Psychiatry ; 20(3): 377-87, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25600109

ABSTRACT

Somatostatin (SST) deficits are common pathological features in depression and other neurological disorders with mood disturbances, but little is known about the contribution of SST deficits to mood symptoms or causes of these deficits. Here we show that mice lacking SST (Sst(KO)) exhibit elevated behavioral emotionality, high basal plasma corticosterone and reduced gene expression of Bdnf, Cortistatin and Gad67, together recapitulating behavioral, neuroendocrine and molecular features of human depression. Studies in Sst(KO) and heterozygous (Sst(HZ)) mice show that elevated corticosterone is not sufficient to reproduce the behavioral phenotype, suggesting a putative role for Sst cell-specific molecular changes. Using laser capture microdissection, we show that cortical SST-positive interneurons display significantly greater transcriptome deregulations after chronic stress compared with pyramidal neurons. Protein translation through eukaryotic initiation factor 2 (EIF2) signaling, a pathway previously implicated in neurodegenerative diseases, was most affected and suppressed in stress-exposed SST neurons. We then show that activating EIF2 signaling through EIF2 kinase inhibition mitigated stress-induced behavioral emotionality in mice. Taken together, our data suggest that (1) low SST has a causal role in mood-related phenotypes, (2) deregulated EIF2-mediated protein translation may represent a mechanism for vulnerability of SST neurons and (3) that global EIF2 signaling has antidepressant/anxiolytic potential.


Subject(s)
Gyrus Cinguli/pathology , Mood Disorders/genetics , Mood Disorders/pathology , Somatostatin/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/blood , Disease Models, Animal , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Indoles/pharmacology , Indoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mood Disorders/blood , Mood Disorders/etiology , Neurons/metabolism , Signal Transduction/genetics , Somatostatin/genetics , Stress, Psychological/blood , Stress, Psychological/complications , Stress, Psychological/genetics , Transcriptome/genetics
7.
Mol Psychiatry ; 20(5): 594-601, 2015 May.
Article in English | MEDLINE | ID: mdl-25092249

ABSTRACT

Cognitive impairment is highly prevalent among individuals with late-life depression (LLD) and tends to persist even after successful treatment. The biological mechanisms underlying cognitive impairment in LLD are complex and likely involve abnormalities in multiple pathways, or 'cascades,' reflected in specific biomarkers. Our aim was to evaluate peripheral (blood-based) evidence for biological pathways associated with cognitive impairment in older adults with LLD. To this end, we used a data-driven comprehensive proteomic analysis (multiplex immunoassay including 242 proteins), along with measures of structural brain abnormalities (gray matter atrophy and white matter hyperintensity volume via magnetic resonance imaging), and brain amyloid-ß (Aß) deposition (PiB-positron emission tomography). We analyzed data from 80 older adults with remitted major depression (36 with mild cognitive impairment (LLD+MCI) and 44 with normal cognitive (LLD+NC)) function. LLD+MCI was associated with differential expression of 24 proteins (P<0.05 and q-value <0.30) related mainly to the regulation of immune-inflammatory activity, intracellular signaling, cell survival and protein and lipid homeostasis. Individuals with LLD+MCI also showed greater white matter hyperintensity burden compared with LLD+NC (P=0.015). We observed no differences in gray matter volume or brain Aß deposition between groups. Machine learning analysis showed that a group of three proteins (Apo AI, IL-12 and stem cell factor) yielded accuracy of 81.3%, sensitivity of 75% and specificity of 86.4% in discriminating participants with MCI from those with NC function (with an averaged cross-validation accuracy of 76.3%, sensitivity of 69.4% and specificity of 81.8% with nested cross-validation considering the model selection bias). Cognitive impairment in LLD seems to be related to greater cerebrovascular disease along with abnormalities in immune-inflammatory control, cell survival, intracellular signaling, protein and lipid homeostasis, and clotting processes. These results suggest that individuals with LLD and cognitive impairment may be more vulnerable to accelerated brain aging and shed light on possible mediators of their elevated risk for progression to dementia.


Subject(s)
Biomarkers/blood , Brain/pathology , Cognition Disorders/etiology , Depression , Proteins/metabolism , Aged , Aged, 80 and over , Aniline Compounds , Benzothiazoles/pharmacokinetics , Brain/diagnostic imaging , Depression/blood , Depression/complications , Depression/pathology , Female , Humans , Image Processing, Computer-Assisted , Machine Learning , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Positron-Emission Tomography , Proteomics/methods , Psychiatric Status Rating Scales , Thiazoles
9.
Genes Brain Behav ; 13(1): 13-24, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24320616

ABSTRACT

In a research environment dominated by reductionist approaches to brain disease mechanisms, gene network analysis provides a complementary framework in which to tackle the complex dysregulations that occur in neuropsychiatric and other neurological disorders. Gene-gene expression correlations are a common source of molecular networks because they can be extracted from high-dimensional disease data and encapsulate the activity of multiple regulatory systems. However, the analysis of gene coexpression patterns is often treated as a mechanistic black box, in which looming 'hub genes' direct cellular networks, and where other features are obscured. By examining the biophysical bases of coexpression and gene regulatory changes that occur in disease, recent studies suggest it is possible to use coexpression networks as a multi-omic screening procedure to generate novel hypotheses for disease mechanisms. Because technical processing steps can affect the outcome and interpretation of coexpression networks, we examine the assumptions and alternatives to common patterns of coexpression analysis and discuss additional topics such as acceptable datasets for coexpression analysis, the robust identification of modules, disease-related prioritization of genes and molecular systems and network meta-analysis. To accelerate coexpression research beyond modules and hubs, we highlight some emerging directions for coexpression network research that are especially relevant to complex brain disease, including the centrality-lethality relationship, integration with machine learning approaches and network pharmacology.


Subject(s)
Gene Regulatory Networks , Nervous System Diseases/genetics , Transcription, Genetic , Animals , Humans
10.
Anal Biochem ; 430(1): 83-91, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22902804

ABSTRACT

The cell division cycle 25 phosphatases (CDC25s) are key regulators of the physiological cell cycle progression. Their overexpression has been reported in a significant number of cancers, and their inhibition appears to be an interesting strategy for treatments. We propose here a rapid screening test allowing the detection of reversible and irreversible CDC25A and -C inhibitors. The test is based on the incubation of the candidate molecules with the human CDC25 proteins followed by an ultrafiltration step. The retentate is then directly analyzed by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry (MALDI-TOFMS) to detect reversible inhibitors or submitted to peptide mass fingerprint (PMF) analysis to reveal irreversible inhibitors covalently bound to the protein active site. After its validation, the protocol is applied to the detection of a novel candidate inhibitor of CDC25s named SV37. The screening procedure, as well as the preliminary biological results, demonstrates that this compound behaves as a reversible inhibitor.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , cdc25 Phosphatases/antagonists & inhibitors , Amino Acid Sequence , Humans , Molecular Sequence Data , cdc25 Phosphatases/chemistry
11.
Transl Psychiatry ; 2: e109, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22832953

ABSTRACT

Altered glial structure and function is implicated in several major mental illnesses and increasing evidence specifically links changes in oligodendrocytes with disrupted mood regulation. Low density and reduced expression of oligodendrocyte-specific gene transcripts in postmortem human subjects points toward decreased oligodendrocyte function in most of the major mental illnesses. Similar features are observed in rodent models of stress-induced depressive-like phenotypes, such as the unpredictable chronic mild stress and chronic corticosterone exposure, suggesting an effect downstream from stress. However, whether oligodendrocyte changes are a causal component of psychiatric phenotypes is not known. Traditional views that identify oligodendrocytes solely as nonfunctional support cells are being challenged, and recent studies suggest a more dynamic role for oligodendrocytes in neuronal functioning than previously considered, with the region adjacent to the node of Ranvier (i.e., paranode) considered a critical region of glial-neuronal interaction. Here, we briefly review the current knowledge regarding oligodendrocyte disruptions in psychiatric disorders and related animal models, with a focus on major depression. We then highlight several rodent studies, which suggest that alterations in oligodendrocyte structure and function can produce behavioral changes that are informative of mood regulatory mechanisms. Together, these studies suggest a model, whereby impaired oligodendrocyte and possibly paranode structure and function can impact neural circuitry, leading to downstream effects related to emotionality in rodents, and potentially to mood regulation in human psychiatric disorders.


Subject(s)
Affect/physiology , Alzheimer Disease/physiopathology , Depressive Disorder, Major/physiopathology , Oligodendroglia/physiology , Schizophrenia/physiopathology , Alzheimer Disease/genetics , Animals , Astrocytes/physiology , Brain/pathology , Brain/physiopathology , Cell Communication/physiology , Cell Count , Demyelinating Diseases/physiopathology , Depressive Disorder, Major/genetics , Disease Models, Animal , Humans , Magnetic Resonance Imaging , Mice , Models, Neurological , Nerve Net/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurons/physiology , Ranvier's Nodes/physiology , Schizophrenia/genetics
12.
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
13.
Transl Psychiatry ; 1: e42, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-22832658

ABSTRACT

Altered oligodendrocyte structure and function is implicated in major psychiatric illnesses, including low cell number and reduced oligodendrocyte-specific gene expression in major depressive disorder (MDD). These features are also observed in the unpredictable chronic mild stress (UCMS) rodent model of the illness, suggesting that they are consequential to environmental precipitants; however, whether oligodendrocyte changes contribute causally to low emotionality is unknown. Focusing on 2'-3'-cyclic nucleotide 3'-phosphodiesterase (Cnp1), a crucial component of axoglial communication dysregulated in the amygdala of MDD subjects and UCMS-exposed mice, we show that altered oligodendrocyte integrity can have an unexpected functional role in affect regulation. Mice lacking Cnp1 (knockout, KO) displayed decreased anxiety- and depressive-like symptoms (i.e., low emotionality) compared with wild-type animals, a phenotypic difference that increased with age (3-9 months). This phenotype was accompanied by increased motor activity, but was evident before neurodegenerative-associated motor coordination deficits (≤ 9-12 months). Notably, Cnp1(KO) mice were less vulnerable to developing a depressive-like syndrome after either UCMS or chronic corticosterone exposure. Cnp1(KO) mice also displayed reduced fear expression during extinction, despite normal amygdala c-Fos induction after acute stress, together implicating dysfunction of an amygdala-related neural network, and consistent with proposed mechanisms for stress resiliency. However, the Cnp1(KO) behavioral phenotype was also accompanied by massive upregulation of oligodendrocyte- and immune-related genes in the basolateral amygdala, suggesting an attempt at functional compensation. Together, we demonstrate that the lack of oligodendrocyte-specific Cnp1 leads to resilient emotionality. However, combined with substantial molecular changes and late-onset neurodegeneration, these results suggest the low Cnp1 seen in MDD may cause unsustainable and maladaptive molecular compensations contributing to the disease pathophysiology.


Subject(s)
2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , Depressive Disorder, Major/genetics , Emotions/physiology , Oligodendroglia/physiology , Stress, Psychological/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/deficiency , Animals , Chronic Disease , Cohort Studies , Depressive Disorder, Major/psychology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Predictive Value of Tests , Random Allocation
15.
Mol Psychiatry ; 12(11): 1042-56, 975, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17420766

ABSTRACT

Normal aging of the brain differs from pathological conditions and is associated with increased risk for psychiatric and neurological disorders. In addition to its role in the etiology and treatment of mood disorders, altered serotonin (5-HT) signaling is considered a contributing factor to aging; however, no causative role has been identified in aging. We hypothesized that a deregulation of the 5-HT system would reveal its contribution to age-related processes and investigated behavioral and molecular changes throughout adult life in mice lacking the regulatory presynaptic 5-HT(1B) receptor (5-HT(1B)R), a candidate gene for 5-HT-mediated age-related functions. We show that the lack of 5-HT(1B)R (Htr1b(KO) mice) induced an early age-related motor decline and resulted in decreased longevity. Analysis of life-long transcriptome changes revealed an early and global shift of the gene expression signature of aging in the brain of Htr1b(KO) mice. Moreover, molecular changes reached an apparent maximum effect at 18-months in Htr1b(KO) mice, corresponding to the onset of early death in that group. A comparative analysis with our previous characterization of aging in the human brain revealed a phylogenetic conservation of age-effect from mice to humans, and confirmed the early onset of molecular aging in Htr1b(KO) mice. Potential mechanisms appear independent of known central mechanisms (Bdnf, inflammation), but may include interactions with previously identified age-related systems (IGF-1, sirtuins). In summary, our findings suggest that the onset of age-related events can be influenced by altered 5-HT function, thus identifying 5-HT as a modulator of brain aging, and suggesting age-related consequences to chronic manipulation of 5-HT.


Subject(s)
Aging/physiology , Gene Expression Regulation/genetics , Gene Expression/genetics , Motor Activity/genetics , Receptor, Serotonin, 5-HT1B/deficiency , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal/physiology , Dopamine Plasma Membrane Transport Proteins/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Hand Strength/physiology , In Situ Hybridization , Maze Learning/physiology , Mice , Mice, Knockout , Microarray Analysis/methods , Reaction Time/physiology , Receptor, Serotonin, 5-HT1B/genetics , Survival Analysis
16.
Neuroscience ; 137(1): 177-86, 2006.
Article in English | MEDLINE | ID: mdl-16309847

ABSTRACT

There has been increasing interest in functional heterogeneity along the septotemporal, dorsal-ventral (D-V) axis of the hippocampus. Although anatomical connectivity and lesion studies point to discrete roles for these sub-regions, the contribution of differential gene expression across this axis has not been systematically studied. Here we present findings from an Affymetrix microarray screen aimed at identifying genes in the CA1 region of the adult murine hippocampus that show significant differential expression along the D-V axis. Our results indicate that the vast majority of monitored genes (>90%) had tissue expression levels that differed by less than 20% between regions, while less than 0.1% of genes had expression levels that varied more than three-fold by sub-region. Only 23 probes showed a CA1 dorsoventral signal intensity ratio greater than three: 18 enriched dorsally and five enriched ventrally. Probes with the greatest difference in expression levels represent a range of genes with known functions in patterning and signaling, as well as genes without known function. Selective screening with digoxigenin-labeled in situ hybridization confirms the existence of CA1 sub-regionalized expression, with some genes exhibiting a graded expression pattern across the D-V axis, and others restricted to a discrete region. Our findings demonstrate that there are gene expression differences across the D-V axis of the adult murine hippocampus within traditionally recognized cytoarchitecturally defined boundaries. Combined with the previously recognized differences in connectivity and results from lesion studies, our data further confirm the existence of functional heterogeneity along the D-V axis.


Subject(s)
Gene Expression Profiling , Hippocampus/anatomy & histology , Hippocampus/physiology , Oligonucleotide Array Sequence Analysis , Animals , In Situ Hybridization , Male , Mice
17.
Behav Pharmacol ; 12(6-7): 429-38, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11742136

ABSTRACT

The serotonin1A (5-HT1A) receptor has been under intense investigation, mostly due to its putative role in both the etiology and therapeutic treatments of depression and anxiety-related behaviors. However, the exact contribution of this receptor to normal brain physiology and disease processes remains poorly understood, due to a complex expression pattern and multiple functions. Recent development in genetic and genomic approaches allows not only for more refined functional dissection, but also for probing large gene databases for unknown gene product interactions. Here, we describe an experimental approach that is based on a combination of regional and temporal genetic manipulations of the 5-HT1A receptor with large-scale gene expression profiling to attempt to untangle the distinct roles for this receptor in particular brain regions, as well as to identify molecular partners that mediate its function. In turn, new leads for understanding mechanisms of anxiety, depression and their pharmacological treatments may be generated.


Subject(s)
Anxiety Disorders/genetics , Depressive Disorder/genetics , Receptors, Serotonin/genetics , Animals , Anxiety Disorders/physiopathology , Brain/physiopathology , Brain Mapping , Depressive Disorder/physiopathology , Gene Expression Regulation/physiology , Humans , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Serotonin/physiology , Receptors, Serotonin, 5-HT1
18.
Eur Neuropsychopharmacol ; 11(6): 413-21, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11704418

ABSTRACT

Recent technological advances in genetic manipulations and DNA microarrays are profoundly altering the landscape of biological research, offering opportunities to investigate biological questions that were only dreamed of a few years ago. With this revolution comes the hope of being able to tackle some of the more arduous challenges that the central nervous system has presented to the research community. Specifically, a major goal in the study of neuropsychiatric disorders has been to identify underlying mechanisms of brain dysfunction with the expectation that these insights may allow a better diagnosis, prevention and effective treatments for these disorders. For the most part, treatments of these disorders have relied on serendipitous discovery of pharmacological entities with therapeutic efficacy, while the causes of the disorders have remained unknown. The serotonin system, and the serotonin(1A) (5-HT(1A)) receptor in particular, have been under intense investigation, mostly due to the fact that serotonergic drugs that directly or indirectly affect the 5-HT(1A) receptor, are effective therapeutic agents in treating patients with various neuropsychiatric disorders, including anxiety and depression. Genetic deletion of the receptor in mouse results in increased anxiety, thus supporting an active role for this receptor in mood regulation. However, the analysis of genetic deletion experiments can be confounded by hidden developmental roles of the missing receptor, by adaptive compensatory mechanisms, as well by the fact that the genes or gene products that are responsible for the cellular and molecular aspects of the phenotype may be several steps removed from the genetic intervention. Here, we present a combined methodological approach of tissue specific and conditional genetic manipulations, with large-scale search for altered gene expression, as an experimental framework to investigate the role of genes with complex functions and/or complex expression patterns. The 5-HT(1A) receptor is used as a model of gene product with complex functions and distributions, and as a prototypical system to which these new genetic approaches are currently being applied.


Subject(s)
Mood Disorders/genetics , Nucleic Acid Hybridization/methods , Animals , Humans , Mice , Mice, Knockout , Nucleic Acid Hybridization/genetics , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT1
19.
Brain Res ; 902(1): 11-7, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11376590

ABSTRACT

An involvement of serotonin (5-HT) 1A receptors in the etiology of psychiatric disorders has been suggested. Hypo-responsiveness of the 5-HT1A receptor is linked to anxiety and constitutive deletion of the 5-HT1A receptor produces anxiety-like behaviors in the mouse. Evidence that 5-HT1A receptor inactivation increases the therapeutic effects of antidepressants has also been presented. The present studies used in vivo microdialysis and homologous recombination techniques to examine the contribution of 5-HT1A autoreceptors to these effects. Basal and fluoxetine-evoked extracellular concentrations of 5-HT were quantified in the striatum, a projection area of dorsal raphe neurons (DRN), of wild-type (WT) and 5-HT1A receptor knock out (KO) mice. The density of 5-HT transporters was also determined. Basal 5-HT concentrations did not differ in WT and KO mice. Fluoxetine (10 mg/kg) increased 5-HT concentrations in both genotypes. This increase was, however, 2-fold greater in KO mice. In contrast, no differences in K(+)-evoked 5-HT concentrations were seen. Similarly, neither basal nor stimulation-evoked DA differed across genotype. Autoradiography revealed no differences between genotype in the density of 5-HT transporters or post-synaptic 5-HT2A receptors, an index of 5-HT neuronal activity. These experiments demonstrate that, under basal and KCl stimulated conditions, adaptive mechanisms in the 5-HT system compensate for the lack of 5-HT1A autoreceptor regulation of DRN. Furthermore, they suggest that the absence of release-regulating 5-HT1A autoreceptors in the DRN can not account for the anxiety phenotype of KO mice. The enhanced response to fluoxetine in KO mice is consistent with pharmacological studies and suggests that adaptive mechanisms that occur in response to 5-HT1A receptor deletion are insufficient to oppose increases in 5-HT concentrations produced by acute inhibition of the 5-HT transporter.


Subject(s)
Brain/drug effects , Fluoxetine/pharmacology , Membrane Transport Proteins , Nerve Tissue Proteins/physiology , Receptors, Presynaptic/physiology , Receptors, Serotonin/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Brain/metabolism , Carrier Proteins/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Haloperidol/pharmacology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microdialysis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Presynaptic/deficiency , Receptors, Presynaptic/genetics , Receptors, Serotonin/deficiency , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT1 , Serotonin Plasma Membrane Transport Proteins , Spiperone/pharmacology
20.
Proc Natl Acad Sci U S A ; 98(4): 1912-7, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11172050

ABSTRACT

Alterations in serotonin (5-hydroxytriptamine, 5-HT), norepinephrine, and gamma-aminobutyric acid have been linked to the pathophysiology of anxiety and depression, and medications that modulate these neurotransmitters are widely used to treat mood disorders. Recently, the neuropeptide substance P (SP) and its receptor, the neurokinin 1 receptor (NK1R), have been proposed as possible targets for new antidepressant and anxiolytic therapies. However, animal and human studies have so far failed to provide a clear consensus on the role of SP in the modulation of emotional states. Here we show that both genetic disruption and acute pharmacological blockade of the NK1R in mice result in a marked reduction of anxiety and stress-related responses. These behavioral changes are paralleled by an increase in the firing rate of 5-HT neurons in the dorsal raphe nucleus, a major source of serotonergic input to the forebrain. NK1R disruption also results in a selective desensitization of 5-HT1A inhibitory autoreceptors, which resembles the effect of sustained antidepressant treatment. Together these results indicate that the SP system powerfully modulates anxiety and suggest that this effect is at least in part mediated by changes in the 5-HT system.


Subject(s)
Anxiety/physiopathology , Receptors, Neurokinin-1/physiology , Serotonin/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Behavior, Animal , Corticosterone/blood , Diazepam/pharmacology , Indoles/pharmacology , Isoindoles , Male , Mice , Mice, Knockout , Neurokinin-1 Receptor Antagonists , Receptors, Neurokinin-1/genetics , Receptors, Serotonin/physiology , Receptors, Serotonin, 5-HT1 , Serotonin Agents/pharmacology , Serotonin Receptor Agonists/pharmacology , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...