Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Neuropsychopharmacology ; 41(4): 1128-37, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26289144

ABSTRACT

The medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) are both integral components of the corticobasal ganglia-thalamic circuitry that regulates addiction-related behaviors. However, the role of afferent inputs from mPFC to NAc in these behaviors is unclear. To address this, we used a Cre-recombinase-dependent viral vector approach to express G(i/o)-coupled DREADDs (designer receptors exclusively activated by designer drugs) selectively in mPFC neurons projecting to the NAc and examined the consequences of attenuating activity of these neurons on the induction of amphetamine sensitization and on drug taking and drug seeking during cocaine self-administration. Surprisingly, decreasing mPFC afferent activity to the NAc only transiently reduced locomotor sensitization and had no effect on drug taking during cocaine self-administration. However, inhibiting corticostriatal afferent activity during sensitization subsequently enhanced conditioned responding. In addition, this manipulation during drug self-administration resulted in slower rates of extinction and increased responding during drug prime-induced reinstatement-an effect that was normalized by inhibiting these corticostriatal afferents immediately before the drug prime. These results suggest that dampening cortical control over the NAc during drug exposure may lead to long-term changes in the ability of drugs and associated stimuli to drive behavior that has important implications for guiding treatments to prevent relapse.


Subject(s)
Amphetamine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Animals , Cues , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Genetic Vectors , Male , Motor Activity/drug effects , Neural Pathways/drug effects , Neural Pathways/physiology , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Rats, Long-Evans , Rats, Sprague-Dawley , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/physiology , Self Administration
2.
Neuroscience ; 299: 97-106, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-25934037

ABSTRACT

Medium spiny neurons (MSNs) constitute 95% of neurons in the dorsal striatum subdivided into direct (striatonigral) and indirect (striatopallidal) pathways. Whereas D1 and D2 receptors and several neuropeptides, including dynorphin and enkephalin, are differentially expressed in these neurons, 5-hydroxytryptamine 6 receptors (5-HT6) are expressed in both pathways. Previous results demonstrate that concurrent 5-HT6 receptor overexpression in MSNs of both pathways in the dorsomedial striatum (DMS) interferes with instrumental learning and that 5-HT6 overexpression in the dorsolateral striatum (DLS) relieves rats from inflexible habitual behaviors. We hypothesized that 5-HT6 receptor-mediated co-activation of both pathways interferes with the differential activation/inhibition of direct/indirect pathways by dopamine. To test this idea, we cloned novel viral vectors to selectively overexpress 5-HT6 receptors in direct or indirect pathway MSNs to deconstruct their role in modulating instrumental learning and habitual responding. We found that increasing 5-HT6 receptor expression in either direct or indirect pathway MSNs of the posterior DMS selectively enhanced or impaired initial acquisition of a discrete instrumental learning task respectively, though all rats were ultimately able to learn the task. In a separate set of experiments, 5-HT6 receptor overexpression in indirect pathway MSNs of the DLS facilitated behavioral flexibility in rats overtrained on a repetitive pressing task using a variable interval schedule of reinforcement, during an omission contingency training session and subsequent probe testing. Together these findings further the notion that 5-HT6 signaling causes balanced activation of opposing MSN pathways by serotonin in sub-regions of the dorsal striatum allowing for more reflective modalities of behavior.


Subject(s)
Conditioning, Operant/physiology , Neostriatum/physiology , Neurons/physiology , Receptors, Serotonin/physiology , Animals , Dynorphins/genetics , Enkephalins/genetics , Genetic Vectors , Male , Neostriatum/metabolism , Neostriatum/virology , Neurons/metabolism , Neurons/virology , Protein Precursors/genetics , Rats , Rats, Long-Evans , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism
3.
Neuroscience ; 298: 436-47, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25907441

ABSTRACT

Located in the nerve terminals of serotonergic neurons, 5-HT1B autoreceptors are poised to modulate synaptic 5-HT levels with precise temporal and spatial control, and play an important role in various emotional behaviors. This study characterized two novel, complementary viral vector strategies to investigate the contribution of 5-HT1B autoreceptors to fear expression, displayed as freezing, during contextual fear conditioning. Increased expression of 5-HT1B autoreceptors throughout the brain significantly decreased fear expression in both wild-type (WT) and 5-HT1B knockout (1BKO) mice when receptor levels were increased with a cell-type-specific herpes simplex virus (HSV) vector injected into the dorsal raphe nucleus (DRN). Additional studies used an intersectional viral vector strategy, in which an adeno-associated virus containing a double-floxed inverted sequence for the 5-HT1B receptor (AAV-DIO-1B) was combined with the retrogradely transported canine adenovirus-2 expressing Cre (CAV-Cre) in order to increase 5-HT1B autoreceptor expression only in neurons projecting from the DRN to the amygdala. Surprisingly, selective expression of 5-HT1B autoreceptors in just this circuit led to an increase in fear expression in WT, but not 1BKO, mice. These results suggest that activation of 5-HT1B autoreceptors throughout the brain may have an overall effect of attenuating fear expression, but activation of subsets of 5-HT1B autoreceptors in particular brain regions, reflecting distinct projections of serotonergic neurons from the DRN, may have disparate contributions to the ultimate response.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Conditioning, Psychological/physiology , Fear , Receptor, Serotonin, 5-HT1B/metabolism , Analysis of Variance , Animals , Dependovirus/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Serotonin, 5-HT1B/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Time Factors , Transduction, Genetic , Tryptophan Hydroxylase/metabolism
4.
Neuroscience ; 176: 120-31, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21182901

ABSTRACT

Previous studies showed that chronic estrogen treatment increases tryptophan hydroxylase-2 (TpH2) mRNA in the caudal dorsal raphe nucleus (DRN), and this increase was associated with decreased anxiety. The present study explored the interaction of estrogen and targeted, bidirectional manipulation of TpH2 expression in the caudal DRN by knockdown or viral overexpression, to decrease or increase tryptophan hydroxylase expression respectively, on anxiety behavior. Rats were ovariectomized and replaced with empty or estradiol capsules (OVX, OVX/E, respectively). Animals received microinfusions of either antisense TpH2 or control morpholino oligonucleotides into caudal DRN and were later tested in the open field test. A separate group of animals were microinfused with TpH2-GFP or GFP-only herpes simplex viral vectors into caudal DRN and tested in the open field. The bidirectional impact of manipulations on TpH2 expression was confirmed using a combination of quantitative protein and mRNA measurements; TpH2 expression changes were limited to discrete subregions of DRN that were targeted by the manipulations. Estradiol decreased anxiety in all behavioral measures. In the OVX/E group, TpH2 knockdown significantly decreased time spent in the center of the open field, but not in the OVX group, suggesting that TpH2 knockdown reduced the anxiolytic effects of estrogen. Conversely, TpH2 overexpression in the OVX group mimicked the effects of estrogen, as measured by increased time spent in the center of the open field. These results suggest that estrogen and TpH2 in the caudal DRN have a critical interaction in regulating anxiety-like behavior.


Subject(s)
Anxiety Disorders/metabolism , Anxiety/metabolism , Behavior, Animal/physiology , Estradiol/metabolism , Raphe Nuclei/metabolism , Tryptophan Hydroxylase/metabolism , Animals , Blotting, Western , Female , Gene Knockdown Techniques , Immunohistochemistry , In Situ Hybridization , Male , Ovariectomy , Rats , Rats, Sprague-Dawley
5.
Neuroscience ; 158(2): 456-64, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19049819

ABSTRACT

We have recently shown that estrogen decreases anxiety and increases expression of tryptophan hydroxylase-2 (TPH2), the rate-limiting enzyme for 5-HT synthesis. However, the effects of estrogen on 5-HT release and reuptake may also affect the overall availability of 5-HT in the forebrain. Estrogen has been previously shown to have no effect on the inhibitory 5-HT 1A autoreceptor (5-HT(1A)) in the rat dorsal raphe nuclei (DRN); however the regulation of the inhibitory 5-HT 1B autoreceptor (5-HT(1B)) in the midbrain raphe by estrogen has not yet been investigated. Therefore, we examined the effects of estrogen on 5-HT(1B) mRNA in the rat DRN, focusing on specific subregions, and whether 5-HT(1B) mRNA levels correlated with TPH2 mRNA levels and with anxiety-like behavior. Ovariectomized rats were treated for 2 weeks with estrogen or placebo, exposed to the open field test, and 5-HT(1A) and 5-HT(1B) mRNA was quantified by in situ hybridization histochemistry. Estrogen had no effect on 5HT(1A) mRNA in any of the DRN subregions examined, confirming a previous report. In contrast, estrogen selectively decreased 5-HT(1B) mRNA in the mid-ventromedial subregion of the DRN, where 5-HT(1B) mRNA was associated with higher anxiety-like behavior and inversely correlated with TPH2 mRNA levels. These results suggest that estrogen may reduce 5-HT(1B) autoreceptor and increase TPH2 synthesis in a coordinated fashion, thereby increasing the capacity for 5-HT synthesis and release in distinct forebrain regions that modulate specific components of anxiety behavior.


Subject(s)
Down-Regulation/drug effects , Estrogens/pharmacology , Exploratory Behavior/physiology , RNA, Messenger/metabolism , Raphe Nuclei/drug effects , Receptor, Serotonin, 5-HT1B/genetics , Animals , Exploratory Behavior/drug effects , Female , Ovariectomy/methods , Raphe Nuclei/anatomy & histology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Statistics as Topic , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
6.
Neuroscience ; 146(4): 1888-905, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17467184

ABSTRACT

Corticotropin releasing factor (CRF) family peptides play key roles in integrating neural responses to stress. Both major CRF receptors have been pharmacologically identified in the dorsal raphe nucleus (DRN), a stress sensitive and internally heterogeneous nucleus supplying many forebrain regions with serotonergic input. Despite the involvement of chronic stress and serotonergic dysfunction in human mood and anxiety disorders, little is known about the effects of chronic CRF receptor activation on the DRN. We infused ovine CRF (1 ng/h), urocortin II (UCNII, 1 ng/h), or vehicle alone into rat DRN over 6 days. During infusion, animals were allowed to freely explore an open field for 15 min on each of 2 days, with the addition of a novel object on the second day. Following behavioral testing, 5-HT1A, 5-HT1B, 5-HT transporter (SERT), and tryptophan hydroxylase-2 (Tph2) expression was examined through the DRN by in situ hybridization. Ovine CRF infusion resulted in significantly decreased novel object touches, climbs, as well as increased latency to first novel object contact. UCNII had a similar but less dramatic effect, decreasing only climbing behavior. Both ovine CRF and UCNII blunted the decrease in corner time expected on re-exposure to the open field. Both peptides also produced regionally specific changes in gene expression: 5-HT1A expression was increased 30% in the mid-rostral ventromedial DRN, while SERT was decreased by 30% in the mid-caudal shell dorsomedial DRN. There also appeared to be a shift in the relative level of Tph2 expression between the ventromedial and core dorsomedial DRN at the mid-rostral level. Changes in 5-HT1A, SERT, and relative Tph2 mRNA abundance were correlated with novel object exploration. These findings suggest chronic intra-DRN administration of CRF agonists decreases exploratory behavior, while producing subregionally limited changes in serotonergic gene expression. These studies may be relevant to mechanisms underlying behavioral changes after chronic stress.


Subject(s)
Corticotropin-Releasing Hormone/administration & dosage , Exploratory Behavior/drug effects , Raphe Nuclei/drug effects , Receptors, Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Tryptophan Hydroxylase/metabolism , Animals , Behavior, Animal/drug effects , Gene Expression/drug effects , Male , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, Serotonin/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Time Factors , Tryptophan Hydroxylase/genetics , Urocortins
7.
Neuropharmacology ; 52(2): 444-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17059838

ABSTRACT

Serotonin 5-HT(1B) receptors modulate behavioral responses to cocaine, but the effects of cocaine on endogenous 5-HT(1B) receptor expression are not known. Therefore, we examined the effect of binge cocaine administration on 5-HT1B mRNA expression in rat brain. We found that chronic, but not acute, binge cocaine exposure increased 5-HT(1B) mRNA by approximately 80% in nucleus accumbens shell and dorsal striatum. Surprisingly, 5-HT(1B) mRNA was increased in nucleus accumbens shell after chronic vehicle treatment as well, but this effect was driven by animals that were housed with cocaine-treated animals. Thus, 5-HT(1B) mRNA is upregulated by repeated exposure to cocaine and perhaps by social stress as well; both of these factors are relevant to the risk for relapse in cocaine addiction.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Neurons/drug effects , Nucleus Accumbens/cytology , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT1B/genetics , Analysis of Variance , Animals , Body Weight , Drug Administration Schedule , Gene Expression Regulation/drug effects , In Situ Hybridization/methods , Male , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B/metabolism
8.
Alcohol ; 38(2): 73-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16839853

ABSTRACT

Serotonin 5-HT(1B) receptors have been linked to alcoholism in humans and alcohol consumption in rodents. We hypothesize that these receptors, which are located on the axon terminals of nucleus accumbens' (NAcc) projection neurons, modulate alcohol reward mechanisms. To test this hypothesis, we measured ethanol consumption by rats that received bilateral microinjections of a viral vector producing 5-HT(1B) overexpression (HA1B/GFP). Other groups received either control (GFP-only) herpes simplex viral vectors into the medial NAcc shell or were handled briefly with no surgery. All animals were housed singly and had continuous access to water, 6% ethanol, and 12% ethanol in their home cages both before and after surgery. There were no differences in the amount or rate of weight gain, amount of food eaten, or total fluid consumed. There were also no differences in the amount of ethanol consumed between groups prior to surgery. However, after surgery, the HA1B/GFP group consumed twice as much ethanol as the other groups. The main effect of total ethanol consumption was significant (p<.05); the control groups did not differ from each other. Whereas there were no between-group differences in 6% ethanol consumption, there was a large increase in the amount of 12% ethanol consumed by the HA1B/GFP-expressing animals compared to the two control groups as well as to their own presurgery intake (p<.05). We hypothesize that increased 5-HT(1B) expression in NAcc led to either greater reward or reduced aversive effects from the 12% ethanol, thereby leading to increased voluntary ethanol consumption.


Subject(s)
Alcohol Drinking/physiopathology , Gene Expression , Nucleus Accumbens/metabolism , Receptor, Serotonin, 5-HT1B/genetics , Transfection , Animals , Eating , Ethanol/administration & dosage , Genetic Vectors , Green Fluorescent Proteins/genetics , Hemagglutinins , Male , Rats , Rats, Long-Evans , Receptor, Serotonin, 5-HT1B/physiology , Recombinant Fusion Proteins , Simplexvirus/genetics , Weight Gain
9.
Pharmacogenomics J ; 5(5): 278-91, 2005.
Article in English | MEDLINE | ID: mdl-16010284

ABSTRACT

In the learned helplessness (LH) animal model of depression, failure to attempt escape from avoidable environmental stress, LH, indicates behavioral despair, whereas nonhelpless (NH) behavior reflects behavioral resilience to the effects of environmental stress. Comparing hippocampal gene expression with large-scale oligonucleotide microarrays, we found that stress-resilient (NH) rats, although behaviorally indistinguishable from controls, showed a distinct gene expression profile compared to LH, sham stressed, and naïve control animals. Genes that were confirmed as differentially expressed in the NH group by quantitative PCR strongly correlated in their levels of expression across all four animal groups. Differential expression could not be confirmed at the protein level. We identified several shared degenerate sequence motifs in the 3' untranslated region (3'UTR) of differentially expressed genes that could be a factor in this tight correlation of expression levels among differentially expressed genes.


Subject(s)
Behavior, Animal/physiology , Depression/genetics , Gene Expression , Helplessness, Learned , Hippocampus/metabolism , Stress, Psychological/genetics , Adaptor Proteins, Vesicular Transport , Animals , Depression/physiopathology , Disease Models, Animal , Electroshock , Gene Expression Profiling , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Stress, Psychological/physiopathology , Untranslated Regions
10.
Physiol Behav ; 84(5): 707-14, 2005 Apr 13.
Article in English | MEDLINE | ID: mdl-15885246

ABSTRACT

Cocaine facilitates dopamine transmission from ventral tegmental area (VTA) neurons that project to nucleus accumbens (NAcc), and previous experiments suggest that serotonin-1B (5-HT1B) receptors are involved in this effect. Specifically, activation of 5-HT1B receptors in VTA during cocaine exposure increases dopamine release in NAcc and enhances cocaine-induced locomotor activity, reward, and reinforcement. Thus, it is reasonable to hypothesize that blocking 5-HT1B activity may have the opposite effect. To investigate this hypothesis, SB 224289, a highly selective 5-HT1B antagonist, was used to block this receptor. In an open field/novel object exploration test, SB 224289 reduced cocaine-induced locomotion. However, SB 224289 also increased anxiety-like behavior, both alone and in combination with cocaine. This experiment gives evidence that 5-HT1B antagonists may reduce some of the behavioral effects of cocaine, but may have negative effects on anxiety as well.


Subject(s)
Anxiety/psychology , Behavior, Animal/drug effects , Cocaine/pharmacology , Piperidones/pharmacology , Receptor, Serotonin, 5-HT1B/drug effects , Serotonin Antagonists/pharmacology , Spiro Compounds/pharmacology , Animals , Dose-Response Relationship, Drug , Environment , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Stimulation, Chemical
11.
J Chem Neuroanat ; 21(1): 63-73, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11173221

ABSTRACT

5-HT(7) receptors are recently identified members of the serotonin receptor family that have moderate to high affinity for several important psychotropic drugs. However, the lack of selective ligands has impeded the study of the brain distribution of these receptors. In this report, we describe the localization of 5-HT(7) receptor in rat forebrain by immunocytochemistry, in situ hybridization of 5-HT(7) mRNA, and functional stimulation of cFOS expression by 5-HT(7) receptor activation. The anatomical localization of 5-HT(7) mRNA in situ hybridization signal. Prominent immunostaining was apparent in numerous sites within the cerebral cortex, hippocampal formation, tenia tecta, thalamus and hypothalamus. 5-HT(7) receptors were detected in suprachiasmatic nucleus by both immunocytochemistry and in situ hybridization. At a microscopic level, both cell bodies and proximal fibers were strongly stained in these regions, suggesting a somatodendritic subcellular distribution. 5-HT(7) receptor-like immunoreactivity was further compared with 5-HT(7) mediated biological function by administering 8-OH-DPAT intracerebroventricular injection (icv)with WAY 100135 (to block 5-HT(1A) receptors) followed by double immunostaining localization of cFos activation and 5-HT(7) receptors. In all regions examined, cFos stimulation and 5-HT(7)-like immunoreactivity colocalized to the same neurons. Furthermore, cFos activation by 8-OH-DPAT was blocked by pimozide--a 5-HT(7) antagonist. Therefore, by using multiple strategies, we were able to localize 5-HT(7) receptors in rat brain unequivocally. The distribution of these receptors is consistent with their involvement in the control of circadian activity and the action of anti-depressants and atypical neuroleptics.


Subject(s)
Brain Chemistry/drug effects , Brain/anatomy & histology , Gene Expression Regulation/drug effects , Genes, fos/drug effects , Receptors, Serotonin/metabolism , Serotonin Receptor Agonists/pharmacology , Animals , Antibodies, Blocking/pharmacology , Blotting, Western , Immunohistochemistry , In Situ Hybridization , Male , RNA, Messenger/biosynthesis , Radioligand Assay , Rats , Rats, Sprague-Dawley
12.
Psychopharmacol Bull ; 35(4): 170-85, 2001.
Article in English | MEDLINE | ID: mdl-12397864

ABSTRACT

5-HT1B receptors are expressed throughout the mammalian central nervous system. These receptors are located in the axon terminals of both serotonergic and nonserotonergic neurons, where they act as inhibitory autoreceptors or heteroreceptors, respectively. 5-HT1B receptors inhibit the release of a range of neurotransmitters, including serotonin, GABA, acetylcholine, and glutamate. These receptors have been difficult to study because of the diversity of their cellular localization and the absence of highly selective agonists and antagonists. There has been accumulating evidence, however, that 5-HT1B receptors modulate drug reinforcement, stress sensitivity, mood, anxiety, and aggression. The general results of a number of studies suggest that reduced 5-HT1B heteroreceptor activity may increase impulsive behaviors, whereas reduced 5-HT1B autoreceptor activity may have an antidepressant-like effect. This review focuses on the evidence from animal studies and human genetics that suggest that 5-HT1B receptors may be involved in the mechanism of action of antidepressants and may become important targets of drug therapy in the future.


Subject(s)
Receptors, Serotonin/drug effects , Serotonin Agents/pharmacology , Aggression/physiology , Animals , Antidepressive Agents/therapeutic use , Humans , Mice , Mice, Knockout , Receptor, Serotonin, 5-HT1B , Receptors, Serotonin/genetics , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Substance-Related Disorders/physiopathology
13.
Brain Res Mol Brain Res ; 82(1-2): 65-73, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11042358

ABSTRACT

The role of mineralocorticoid and glucocorticoid receptors (MR and GR, respectively) in the regulation of serotonin receptors has not been clearly delineated. There is no consensus regarding the regulation of 5-HT(1A) receptors, and corticosteroid regulation of 5-HT(1B) mRNA has not been previously studied. We compared the effects of long-term (two week) adrenalectomy (no MR or GR activation) and several hormone replacement protocols designed to stimulate MR selectively (ALDO), MR and GR (HCT), and continuous MR with cyclical GR activation (SHAM adrenalectomy). 5-HT(1A) and 5-HT(1B) mRNAs were measured by in situ hybridization in hippocampus and raphe nuclei. None of the experimental manipulations altered 5-HT(1B) mRNA levels in the hippocampus or dorsal raphe, and also had no effect on 5-HT(1A) mRNA in dorsal or median raphe. However, 5-HT(1A) mRNA levels were regulated in a complex manner in the different subfields of hippocampus. We conclude that both MR and GR play an integrated role in regulating 5-HT(1A) mRNA levels in hippocampus while having no effect on 5-HT(1B) mRNA levels under these conditions.


Subject(s)
Aldosterone/pharmacology , Gene Expression Regulation/physiology , Hippocampus/metabolism , Receptors, Glucocorticoid/physiology , Receptors, Mineralocorticoid/physiology , Receptors, Serotonin/genetics , Transcription, Genetic/physiology , Adrenalectomy , Animals , Gene Expression Regulation/drug effects , In Situ Hybridization , Male , RNA, Messenger/genetics , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B , Receptors, Serotonin, 5-HT1 , Serotonin/physiology , Transcription, Genetic/drug effects
14.
Neurosci Lett ; 288(3): 236-40, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10889351

ABSTRACT

Clozapine is an atypical antipsychotic with high affinity for several serotonin receptors. This drug causes paradoxical downregulation of 5-hydroxytryptamine(2A) (5-HT)(2A) receptors, but its modulation of other serotonin receptors has not been studied. We examined the effects of clozapine and several other drugs on the regulation of rat 5-HT(6) and 5-HT(7) receptors individually expressed in transfected HeLa cells. Both 5-HT(6) and 5-HT(7) receptor densities (B(max)) were reduced by 5-carboxamidotryptamine, an agonist, and methiothepin, an inverse agonist. Clozapine reduced 5-HT(6) B(max). This suggests that 5-HT(6) receptors are also paradoxically downregulated by the antagonist clozapine. 5-Hydroxytryptamine(7) receptor B(max), on the other hand, was increased by clozapine. Clozapine's modulation of the 5-HT(6) and 5-HT(7) receptor levels may be important in the action of this atypical antipsychotic.


Subject(s)
Clozapine/pharmacology , Down-Regulation/drug effects , HeLa Cells/drug effects , HeLa Cells/metabolism , Receptors, Serotonin/biosynthesis , Serotonin Antagonists/pharmacology , Serotonin/analogs & derivatives , Serotonin/biosynthesis , Up-Regulation/drug effects , Animals , Humans , Methiothepin/pharmacology , Rats , Serotonin/pharmacology , Serotonin Receptor Agonists/pharmacology
15.
J Neurosci Res ; 61(1): 82-7, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861803

ABSTRACT

Serotonin release from dorsal raphe projections in the forebrain is regulated by terminal 5-HT(1B) autoreceptors; dysregulation of these receptors may be involved in the pathophysiology of clinical depression. Using in situ hybridization, we have previously reported that fluoxetine reduces 5-HT(1B) mRNA in rat dorsal raphe nucleus (DRN) in a time-dependent and reversible manner. In this study we examined longer term treatment (8 weeks) with several different serotonin-selective reuptake inhibitors (SSRIs) or a tricyclic antidepressant on 5-HT(1B) mRNA regulation in DRN and hippocampus, and evaluated the stability of these drugs' effects after drug discontinuation. Fluoxetine (5 mg/kg/d), paroxetine (5 mg/kg/d), sertraline (10 mg/kg/d) or nortriptyline (10 mg/kg/d) was administered to rats via subcutaneous osmotic minipumps. Paroxetine and fluoxetine reduced DRN 5-HT(1B) mRNA by 36% and 27%, respectively whereas sertraline had a no significant effect. After 3-14 days of drug washout, DRN 5-HT(1B) mRNA levels in SSRI treated rats were no longer different from control. 5-HT(1B) mRNA levels in hippocampus were not affected by SSRI drugs at any timepoint. Nortriptyline had no significant effect on 5-HT(1B) mRNA in either DRN or hippocampus. These results confirm that SSRI antidepressants reduce presynaptic 5-HT(1B) mRNA selectively, and that this effect is maintained for at least 8 weeks of antidepressant treatment but reverses rapidly after discontinuation. Furthermore, it is possible that washout after chronic antidepressant treatment, that is routinely used in functional assays of autoreceptor action in animal models, may lead to more rapid reversal of biological effects than has previously been thought.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Paroxetine/pharmacology , Raphe Nuclei/physiology , Receptors, Serotonin/genetics , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Autoreceptors/physiology , Fluoxetine/pharmacology , Gene Expression/drug effects , In Situ Hybridization , Male , Nortriptyline/pharmacology , RNA, Messenger/metabolism , Raphe Nuclei/chemistry , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B , Sertraline/pharmacology
16.
Mol Psychiatry ; 4(6): 572-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10578240

ABSTRACT

(+) 3,4-Methylenedioxymethamphetamine (MDMA) is a psychedelic drug of abuse that causes selective degeneration of serotonergic fibers of dorsal raphe neurons that project throughout the forebrain. Previous studies using pharmacological and behavioral approaches suggested that MDMA treatment leads to desensitization of 5-HT1B receptors. We proposed to test whether this occurs by downregulation of 5-HT1B messenger RNA in dorsal raphe, striatum or CA1 hippocampal neurons and/or 5-HT1B binding site density in hippocampus and basal ganglia. In Experiment I, rats were treated with MDMA using several dosing protocols (2.5 or 10 mg kg-1 day-1 s.c. given a single time or twice daily for 4 days). The animals were killed 24 h after the last dose. [3H]-citalopram binding to serotonin transporters in hippocampus was reduced in the high dose protocol, indicating degeneration of forebrain serotonergic fibers. Despite the extensive reduction in serotonergic content, 5-HT1B mRNA did not change from control levels in any region when measured by in situ hybridization. [125I]-Iodocyanopindolol binding to 5-HT1B sites in hippocampus was also not changed. In Experiment II, high dose MDMA had no effect on 5-HT1B mRNA in any brain region either 1 or 14 days after treatment. However, [125I]-iodocyanopindolol binding more than doubled in striatum 1 day after MDMA treatment but returned to control levels by 14 days. This may have been a transient compensation to early neuronal damage caused by MDMA exposure. These results suggest that previously described changes in 5-HT1B function following MDMA treatment involve only posttranscriptional changes in receptor regulation and do not alter 5-HT1B mRNA levels.


Subject(s)
Brain Chemistry/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Serotonin Agents/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Citalopram/pharmacology , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hippocampus/chemistry , Hippocampus/drug effects , In Situ Hybridization , Iodine Radioisotopes , Iodocyanopindolol/pharmacology , RNA, Messenger/metabolism , Radioligand Assay , Raphe Nuclei/chemistry , Raphe Nuclei/drug effects , Rats , Receptor, Serotonin, 5-HT1B , Selective Serotonin Reuptake Inhibitors/pharmacology , Tritium
18.
Neuroreport ; 10(18): 3725-31, 1999 Dec 16.
Article in English | MEDLINE | ID: mdl-10716199

ABSTRACT

Developing in vitro blood-brain barrier (BBB) models that closely mimic the natural state is important for theoretical and practical applications, including drug development. We previously developed an in vitro BBB model based on co-culturing endothelial cells with glia in the presence of flow on hollow fiber tube culture substrates. We now report that this dynamic in vitro BBB (DIV-BBB) can be successfully used to co-culture differentiated serotonergic neurons in the presence of a BBB. These neurons demonstrated fluoxetine-sensitive serotonin (5HT) uptake and depolarization-induced release of [3H]5HT. Our results demonstrate that the DIV-BBB is a suitable model for culturing of neurons in a quasi-physiological microenvironment and in the presence of a high-resistance, stereoselective BBB.


Subject(s)
Blood-Brain Barrier/physiology , Animals , Cerebrovascular Circulation/physiology , Coculture Techniques , Electrophysiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Fluoxetine/pharmacology , Immunohistochemistry , Neuroglia/physiology , Neurons/physiology , Perfusion , Rats , Serotonin/metabolism , Serotonin/pharmacokinetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Tumor Cells, Cultured
19.
Biol Psychiatry ; 41(6): 668-74, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9066990

ABSTRACT

Learned helplessness is a behavioral condition induced by exposure to inescapable stress that models aspects of stress-related disorders including depression and posttraumatic stress disorder, and has been associated with diminished serotonin release in the rat frontal cortex. Our hypothesis was that presynaptic 5-hydroxytryptamine1B (5-HT1B) receptors, which inhibit the synthesis and release of serotonin in nerve terminals, may be increased in learned helplessness. Postsynaptic 5-HT1B mRNA hybridization levels in the hippocampus or frontal cortex were unchanged following induction of learned helplessness; however, presynaptic 5-HT1B mRNA hybridization signal in the dorsal raphe nucleus of helpless rats was 25% higher than control values. There was no change in dorsal raphe serotonin transporter mRNA level. The detection of increased 5-HT1B mRNA levels in the dorsal raphe nucleus suggests an increased capacity to synthesize presynaptic 5-HT1B receptors and could account for diminished serotonin neurotransmission in learned helplessness.


Subject(s)
Helplessness, Learned , RNA, Messenger/biosynthesis , Raphe Nuclei/metabolism , Receptors, Serotonin/biosynthesis , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Densitometry , Hippocampus/cytology , Hippocampus/metabolism , In Situ Hybridization , Male , Pyramidal Cells/metabolism , Rats
20.
Neuropsychopharmacology ; 15(5): 515-22, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8914125

ABSTRACT

In major depression in humans and in animal models of depression, there is a defect in serotonergic neurotransmission that can be relieved by chronic antidepressant treatment. One possibility is that this pathologic state is caused by excessive presynaptic autoreceptor activity in serotonergic neurons, and that antidepressants down-regulate the number of these inhibitory receptors, allowing more normal serotonin release to occur. To evaluate this hypothesis, we measured the effects of the antidepressant fluoxetine on neuronal levels of 5-HT1B receptor mRNA, the putative serotonin terminal autoreceptor in rat brain, and on serotonin transporter mRNA, the direct site of fluoxetine binding. Fluoxetine reduced serotonin transporter mRNA briefly, but this was not sustained after 21 days of treatment. However, fluoxetine reduced dorsal raphe 5-HT1B mRNA levels in a time-dependent and washout-reversible manner. This reduction in 5-HT1B mRNA was specific to dorsal raphe nucleus and was not found in several postsynaptic (nonserotonergic) regions. These results suggest that chronic fluoxetine may increase serotonin release from axonal terminals by down-regulating the messenger RNA coding for presynaptic 5-HT1B autoreceptors while causing only transient effects on serotonin transporter mRNA.


Subject(s)
Carrier Proteins/metabolism , Fluoxetine/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins/metabolism , RNA, Messenger/analysis , Raphe Nuclei/drug effects , Receptors, Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , In Situ Hybridization , Male , Raphe Nuclei/chemistry , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B , Serotonin , Serotonin Plasma Membrane Transport Proteins , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...