Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 54(10): 8263-8277, 2017 12.
Article in English | MEDLINE | ID: mdl-27914010

ABSTRACT

Five-prime repressor element under dual repression binding protein-1 (Freud-1)/CC2D1A is genetically linked to intellectual disability and implicated in neuronal development. Freud-1 represses the serotonin-1A (5-HT1A) receptor gene HTR1A by histone deacetylase (HDAC)-dependent or HDAC-independent mechanisms in 5-HT1A-negative (e.g., HEK-293) or 5-HT1A-expressing cells (SK-N-SH), respectively. To identify the underlying mechanisms, Freud-1-associated proteins were affinity-purified from HEK-293 nuclear extracts and members of the Brg1/SMARCCA chromatin remodeling and Sin3A-HDAC corepressor complexes were identified. Pull-down assays using recombinant proteins showed that Freud-1 interacts directly with the Brg1 carboxyl-terminal domain; interaction with Brg1 required the carboxyl-terminal of Freud-1. Freud-1 complexes in HEK-293 and SK-N-SH cells differed, with low levels of BAF170/SMARCC2 and BAF57/SMARCE1 in HEK-293 cells and low-undetectable BAF155/SMARCC1, Sin3A, and HDAC1/2 in SK-N-SH cells. Similarly, by quantitative chromatin immunoprecipitation, Brg1-BAF170/57 and Sin3A-HDAC complexes were observed at the HTR1A promoter in HEK-293 cells, whereas in SK-N-SH cells, Sin3A-HDAC proteins were not detected. Quantifying 5-HT1A receptor mRNA levels in cells treated with siRNA to Freud-1, Brg1, or both RNAs addressed the functional role of the Freud-1-Brg1 complex. In HEK-293 cells, 5-HT1A receptor mRNA levels were increased only when both Freud-1 and Brg1 were depleted, but in SK-N-SH cells, depletion of either protein upregulated 5-HT1A receptor RNA. Thus, recruitment by Freud-1 of Brg1, BAF155, and Sin3A-HDAC complexes appears to strengthen repression of the HTR1A gene to prevent its expression inappropriate cell types, while recruitment of the Brg1-BAF170/57 complex is permissive to 5-HT1A receptor expression. Alterations in Freud-1-Brg1 interactions in mutants associated with intellectual disability could impair gene repression leading to altered neuronal development.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA Helicases/biosynthesis , DNA-Binding Proteins/biosynthesis , Histone Deacetylases/biosynthesis , Nuclear Proteins/biosynthesis , Receptor, Serotonin, 5-HT1A/biosynthesis , Transcription Factors/biosynthesis , DNA Helicases/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Histone Deacetylases/genetics , Humans , Nuclear Proteins/genetics , Receptor, Serotonin, 5-HT1A/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transcription Factors/genetics
2.
Neurobiol Dis ; 82: 332-341, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26188176

ABSTRACT

The serotonin 1A receptor (5-HT1A), a critical regulator of the brain serotonergic tone, is implicated in major depressive disorder (MDD) where it is often found to be dys-regulated. However, the extent to which stress and antidepressant treatment impact 5-HT1A expression in adults remains unclear. To address this issue, we subjected adult male BALB/c mice to unpredictable chronic mild stress (UCMS) to induce a depression-like phenotype that was reversed by chronic treatment with the antidepressant imipramine. In prefrontal cortex (PFC) and midbrain tissue, UCMS increased 5-HT1A RNA and protein levels, changes that are expected to decrease the brain serotonergic activity. The stress-induced increase in 5-HT1A expression was paralleled by a specific increase in DNA methylation of the conserved -681 CpG promoter site, located within a Sp1-like element. We show that the -681 CpG site is recognized and repressed by Sp4, the predominant neuronal Sp1-like factor and that Sp4-induced repression is attenuated by DNA methylation, despite a stress-induced increase in PFC Sp4 levels. These results indicate that adult life stress induces DNA methylation of a conserved promoter site, antagonizing Sp4 repression to increase 5-HT1A expression. Chronic imipramine treatment fully reversed the UCMS-induced increase in methylation of the -681 CpG site in the PFC but not midbrain of stressed animals and also increased 5-HT1A expression in the PFC of control animals. Incomplete reversal by imipramine of stress-induced changes in 5-HT1A methylation and expression indicates a persistence of stress vulnerability, and that sustained reversal of behavioral impairments may require additional pathways.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , DNA Methylation/drug effects , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Animals , Chronic Disease , Conserved Sequence , CpG Islands , DNA Methylation/physiology , Depressive Disorder/genetics , Disease Models, Animal , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Imipramine/pharmacology , Male , Mice, Inbred BALB C , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/genetics , Stress, Psychological/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
3.
J Biol Chem ; 287(9): 6615-27, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22232550

ABSTRACT

Altered regulation of the serotonin-1A (5-HT1A) receptor gene is implicated in major depression and mood disorders. The functional human 5-HT1A C(-1019)G promoter polymorphism (rs6295), which prevents the binding of Deaf-1/NUDR leading to dysregulation of the receptor, has been associated with major depression. In cell models Deaf-1 displays dual activity, repressing 5-HT1A autoreceptor expression in serotonergic raphe cells while enhancing postsynaptic 5-HT1A heteroreceptor expression in nonserotonergic neurons. A functional Deaf-1 binding site on the mouse 5-HT1A promoter was recognized by Deaf-1 in vitro and in vivo and mediated dual activity of Deaf-1 on 5-HT1A gene transcription. To address regulation by Deaf-1 in vivo, Deaf-1 knock-out mice bred to a C57BL/6 background were compared with wild-type siblings for changes in 5-HT1A RNA and protein by quantitative RT-PCR, in situ hybridization, and immunofluorescence. In the dorsal raphe, Deaf-1 knock-out mice displayed increased 5-HT1A mRNA, protein, and 5-HT1A-positive cell counts but reduced 5-HT levels, whereas other serotonergic markers, such as tryptophan hydroxylase (TPH)- or 5-HT-positive cells and TPH2 RNA levels, were unchanged. By contrast, 5-HT1A mRNA and 5-HT1A-positive cells were reduced in the frontal cortex of Deaf-1-null mice, with no significant change in hippocampal 5-HT1A RNA, protein, or cell counts. The region-specific alterations of brain 5-HT1A gene expression and reduced raphe 5-HT content in Deaf-1(-/-) mice indicate the importance of Deaf-1 in regulation of 5-HT1A gene expression and provide insight into the role of the 5-HT1A G(-1019) allele in reducing serotonergic neurotransmission by derepression of 5-HT1A autoreceptors.


Subject(s)
Autoreceptors/genetics , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT1A/genetics , Serotonin/metabolism , Transcription Factors/genetics , Animals , Autoreceptors/metabolism , DNA-Binding Proteins , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Female , Fluorescent Antibody Technique , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Transcription Factors/metabolism , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
4.
Biol Psychol ; 88(1): 83-93, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21742012

ABSTRACT

Given the cognitive-promoting properties of the nicotinic acetylcholinergic receptor (nAChR) agonist, nicotine, the increased prevalence of smoke-inhaled nicotine in schizophrenia has been interpreted as an attempt to self-correct cognitive deficits, which have been particularly pronounced in the attentional domain. As glutamatergic abnormalities have been implicated in these attentional deficiencies, this study attempted to shed light on the separate and interactive roles of the N-methyl-d-aspartate receptor (NMDAR) and nAChR systems in the modulation of attention by investigating, in healthy volunteers, the separate and combined effects of nicotine and the NMDAR antagonist ketamine on neural and behavioural responses in a sustained attention task. In a randomized, double-blind, placebo controlled study, performance and the P300 event-related brain potential (ERP) in a visual information processing (RVIP) task were examined in 20 smokers and 20 non-smokers (both male and female). Assessment involved intravenous injection of a low subperceptual bolus dose (.04mg/kg) of ketamine or placebo, which was accompanied by acute treatment with nicotine (4mg) or placebo gum. Nicotine-enhanced attentional processing was most evident in nonsmokers, with both performance accuracy and P300 amplitude measures. Ketamine's detrimental effects on these behavioural and electrophysiologic measures were negatively moderated by acute nicotine, the synergistic effects being expressed differently in smokers and nonsmokers. These findings support the view that acute alterations and individual differences in nAChR function can moderate even subtle glutamatergic-driven cognitive deficiencies in schizophrenia and can be important therapeutic targets for treating cognitive impairments in schizophrenia.


Subject(s)
Analgesics/pharmacology , Attention/drug effects , Ketamine/pharmacology , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Adolescent , Adult , Analysis of Variance , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Electroencephalography , Electrooculography , Event-Related Potentials, P300/drug effects , Female , Humans , Male , Middle Aged , Photic Stimulation , Reaction Time/drug effects , Smoking/drug therapy , Smoking/physiopathology , Surveys and Questionnaires , Young Adult
5.
Mol Brain ; 4: 21, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21619616

ABSTRACT

The serotonin-1A (5-HT1A) receptor is among the most abundant and widely distributed 5-HT receptors in the brain, but is also expressed on serotonin neurons as an autoreceptor where it plays a critical role in regulating the activity of the entire serotonin system. Over-expression of the 5-HT1A autoreceptor has been implicated in reducing serotonergic neurotransmission, and is associated with major depression and suicide. Extensive characterization of the transcriptional regulation of the 5-HT1A gene (HTR1A) using cell culture systems has revealed a GC-rich "housekeeping" promoter that non-selectively drives its expression; this is flanked by a series of upstream repressor elements for REST, Freud-1/CC2D1A and Freud-2/CC2D1B factors that not only restrict its expression to neurons, but may also regulate the level of expression of 5-HT1A receptors in various subsets of neurons, including serotonergic neurons. A separate set of allele-specific factors, including Deaf1, Hes1 and Hes5 repress at the HTR1A C(-1019)G (rs6295) polymorphism in serotonergic neurons in culture, as well as in vivo. Pet1, an obligatory enhancer for serotonergic differentiation, has been identified as a potent activator of 5-HT1A autoreceptor expression. Taken together, these results highlight an integrated regulation of 5-HT1A autoreceptors that differs in several aspects from regulation of post-synaptic 5-HT1A receptors, and could be selectively targeted to enhance serotonergic neurotransmission.


Subject(s)
Autoreceptors/genetics , Gene Expression Regulation , Mental Disorders/genetics , Receptor, Serotonin, 5-HT1A/genetics , Transcription, Genetic , Autoreceptors/metabolism , Humans , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism
6.
Neuropsychobiology ; 58(3-4): 187-99, 2008.
Article in English | MEDLINE | ID: mdl-19212134

ABSTRACT

BACKGROUND/AIMS: Cigarette craving is a core symptom of smoking withdrawal, which is more intense and more frequently observed in smokers with depressed mood. Using self-reports and electroencephalographic (EEG) indices of frontal hemispheric asymmetry, which has been shown to be sensitive to mood states, the purpose of this study was to investigate the neural basis of cue-elicited cigarette craving, its variation with experimentally induced depressed mood, and with differences in gender and smoker type. METHODS: Cigarette-cue reactivity was examined in 11 (5 male) regular and 11 (6 male) light smokers in two sessions involving the induction of neutral or depressed mood. RESULTS: Frontal EEG alpha asymmetry changes reflecting left frontal hypoactivation were evident with cigarette-cue exposure, particularly in female smokers. During cigarette-cue exposure, EEG evidenced both decreases and increases in brain state activation, with the latter activational increments also being influenced by depressed mood. Exposure to the cigarette cue, in addition to increasing withdrawal symptoms, increased cravings and negative affect, these latter effects being more evident in female and regular smokers. CONCLUSION: These findings, which appear to provide a physiological basis for 'withdrawal-like' negative affective experiences during craving, are discussed in relation to theories of drug reinforcement and smoking motivation.


Subject(s)
Affect/physiology , Brain/physiopathology , Sex Characteristics , Smoking/psychology , Tobacco Use Disorder/physiopathology , Adult , Cues , Depression/physiopathology , Electroencephalography , Female , Humans , Male , Neuropsychological Tests , Smoking/physiopathology , Substance Withdrawal Syndrome/physiopathology , Surveys and Questionnaires , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...