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

Database
Language
Affiliation country
Publication year range
1.
J Chem Neuroanat ; 132: 102324, 2023 10.
Article in English | MEDLINE | ID: mdl-37557929

ABSTRACT

Advances have been made in recent years in using opioid receptor antagonists as an adjunct therapy to psychotropic medication to reduce debilitating weight gain and metabolic adverse effects associated with in particular second generation antipsychotics. However, it is unknown whether second generation antipsychotics produce a change in opioid receptor expression in the brain. The present study investigated early changes in opioid receptor expression in the female rat hypothalamus, a master controller of hunger and metabolic regulation, after acute treatment with olanzapine, a commonly used second generation antipsychotic. Using quantitative spatial in situ hybridization and receptor autoradiography, expression levels of the three opioid receptors; kappa, mu and delta, were determined at mRNA and protein level, respectively, in the five hypothalamic areas: paraventricular nucleus, arcuate nucleus, ventromedial nucleus, dorsomedial nucleus and lateral hypothalamus. After 48 h of olanzapine treatment at clinically relevant plasma concentration weight gain and food intake changes, and increased plasma glucose were observed in female rats. Olanzapine treatment also led to a significant increase in mu opioid receptor availability in the arcuate nucleus, which contains both satiety and hunger controlling neurons. No other areas showed any opioid receptor expressional changes with olanzapine treatment on neither at mRNA nor protein level. Technical difficulties made it impossible to analyze mRNA levels in the lateral hypothalamus and overall binding of delta opioid receptors. Thus, the present study provided insights in to how olanzapine at clinically relevant plasma levels already at an early stage modulated the opioid system in the hypothalamus.


Subject(s)
Antipsychotic Agents , Receptors, Opioid, mu , Rats , Female , Animals , Olanzapine/pharmacology , Olanzapine/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Antipsychotic Agents/pharmacology , Hypothalamus/metabolism , Receptors, Opioid/metabolism , Feeding Behavior , Weight Gain , RNA, Messenger
2.
J Chem Neuroanat ; 127: 102205, 2023 01.
Article in English | MEDLINE | ID: mdl-36464066

ABSTRACT

Detailed quantification of brain tissue provides a deeper understanding of changes in expression and function. We have created a pipeline to study the detailed expression patterns of the kappa opioid receptor in the rat hypothalamus using high resolution fluorescence microscopy and receptor autoradiography. The workflow involved structured serial sampling of rat hypothalamic nuclei, in situ detection of mRNA and receptor expression, and advanced image analysis. Our results demonstrate how maintaining spatial information can lead to increased understanding of RNA and protein expression. In addition, we show the detailed expression patterns of the kappa opioid receptor in the rat hypothalamus.


Subject(s)
Hypothalamus , Receptors, Opioid, kappa , Rats , Animals , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , RNA, Messenger , Ligands , In Situ Hybridization , Hypothalamus/metabolism , Autoradiography
3.
Brain Res ; 1664: 37-47, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28366617

ABSTRACT

Studies of the antidepressant vortioxetine have demonstrated beneficial effects on cognitive dysfunction associated with depression. To elucidate how vortioxetine modulates neuronal activity during cognitive processing we investigated the effects of vortioxetine (3 and 10mg/kg) in rats performing an auditory oddball (deviant target) task. We investigated neuronal activity in target vs non-target tone responses in vehicle-treated animals using electroencephalographic (EEG) recordings. Furthermore, we characterized task performance and EEG changes in target tone responses of vortioxetine vs controls. Quantification of event-related potentials (ERPs) was supplemented by analyses of spectral power and inter-trial phase-locking. The assessed brain regions included prelimbic cortex, the hippocampus, and thalamus. As compared to correct rejection of non-target tones, correct target tone responses elicited increased EEG power in all regions. Additionally, neuronal synchronization was increased in vehicle-treated rats during both early and late ERP responses to target tones. This indicates a significant consistency of local phases across trials during high attentional load. During early sensory processing, vortioxetine increased both thalamic and frontal synchronized gamma band activity and EEG power in all brain regions measured. Finally, vortioxetine increased the amplitude of late hippocampal P3-like ERPs, the rodent correlate of the human P300 ERP. These findings suggest differential effects of vortioxetine during early sensory registration and late endogenous processing of auditory discrimination. Strengthened P3-like ERP response may relate to the pro-cognitive profile of vortioxetine in rodents. Further investigations are warranted to explore the mechanism by which vortioxetine increases network synchronization during attentive and cognitive processing.


Subject(s)
Antidepressive Agents/administration & dosage , Attention/drug effects , Brain/drug effects , Brain/physiology , Cognition/drug effects , Evoked Potentials, Auditory/drug effects , Piperazines/administration & dosage , Sulfides/administration & dosage , Acoustic Stimulation , Animals , Attention/physiology , Auditory Perception/drug effects , Auditory Perception/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Cognition/physiology , Electroencephalography , Hippocampus/drug effects , Hippocampus/physiology , Male , Rats, Sprague-Dawley , Thalamus/drug effects , Thalamus/physiology , Vortioxetine
4.
J Neurochem ; 142(1): 118-131, 2017 07.
Article in English | MEDLINE | ID: mdl-28407315

ABSTRACT

The metabolism of tryptophan through kynurenine and serotonin pathways is linked to depression. Here, effects of different drugs with antidepressant properties (vortioxetine, fluoxetine, and ketamine) on various tryptophan metabolites in different brain regions and plasma were examined using tandem mass spectrometry (LC-MS/MS), in Flinders Sensitive Line rats, a genetic rat model of depression, and its controls: Flinders Sensitive Line and Sprague-Dawley rats. Protein levels of kynurenine pathway enzymes were measured in the brains and livers of these rat strains. Furthermore, effects of vortioxetine on tryptophan metabolites were assessed in the cortical regions of lupus mice (MRL/MpJ-FasIpr ), a murine model of increased depression-like behavior associated with inflammation. Sustained vortioxetine or fluoxetine (at doses aimed to fully occupy serotonin transporter via food or drinking water for at least 14 days) reduced levels of the excitotoxin quinolinic acid (QUIN) in various brain regions in all rats. Furthermore, chronic vortioxetine reduced levels of QUIN in MRL/MpJ-FasIpr mice. Acute i.p. administration of fluoxetine (10 mg/kg) or vortioxetine (10 mg/kg) led to reduced brain 5-hydroxyindoleacetic acid in Sprague-Dawley rats (2, 4, 6, and 8 h) and a similar trend was evident in Flinders Sensitive Line and Flinders Sensitive Line rats after 4 h. In contrast, single or repeated administration of ketamine (15 mg/kg i.p.) did not induce significant changes in metabolite levels. In conclusion, sustained vortioxetine and fluoxetine administration decreased QUIN independent of species, while ketamine was ineffective. These results support the hypothesis that modulating tryptophan metabolism may be part of the mechanism of action for some antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Depression/metabolism , Depression/psychology , Tryptophan/metabolism , Animals , Brain Chemistry/drug effects , Fluoxetine/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hydroxyindoleacetic Acid/metabolism , Ketamine/pharmacology , Kynurenine/metabolism , Lupus Erythematosus, Systemic/metabolism , Male , Mice , Piperazines/pharmacology , Quinolinic Acid/metabolism , Rats , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Sulfides/pharmacology , Vortioxetine
5.
Eur J Pharmacol ; 786: 29-35, 2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27235984

ABSTRACT

Ketamine is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist that has been shown to induce a rapid antidepressant effect in treatment-resistant patients. Vortioxetine is a multimodal-acting antidepressant that exert its therapeutic activity through serotonin (5-hydroxytryptamine; 5-HT) reuptake inhibition and modulation of several 5-HT receptors. In clinical trials, vortioxetine improves depression symptoms and cognitive dysfunction. Neuroplasticity as well as serotonergic and glutamatergic signaling attain significant roles in depression pathophysiology and antidepressant responses. Here, we investigate the effects of ketamine and vortioxetine on gene expression related to serotonergic and glutamatergic neurotransmission as well as neuroplasticity and compare them to those of the selective serotonin reuptake inhibitor fluoxetine. Rats were injected with fluoxetine (10mg/kg), ketamine (15mg/kg), or vortioxetine (10mg/kg) at 2, 8, 12, or 27h prior to harvesting of the frontal cortex and hippocampus. mRNA levels were measured by real-time quantitative polymerase chain reaction (qPCR). The main finding was that vortioxetine enhanced plasticity-related gene expression (Mtor, Mglur1, Pkcα, Homer3, Spinophilin, and Synapsin3) in the frontal cortex at 8h after a single dose. Ingenuity pathway analysis of this subset of data identified a biological network that was engaged by vortioxetine and is plausibly associated with neuroplasticity. Transcript levels had returned to baseline levels 12h after injection. Only minor effects on gene expression were found for ketamine or fluoxetine. In conclusion, acute vortioxetine, but not fluoxetine or ketamine, transiently increased plasticity-related gene expression in the frontal cortex. These effects may be ascribed to the direct 5-HT receptor activities of vortioxetine.


Subject(s)
Antidepressive Agents/pharmacology , Frontal Lobe/drug effects , Gene Expression Regulation/drug effects , Neuronal Plasticity/genetics , Piperazines/pharmacology , Sulfides/pharmacology , Animals , Antidepressive Agents/administration & dosage , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , Frontal Lobe/metabolism , Frontal Lobe/physiology , Glutamic Acid/metabolism , Ketamine/pharmacology , Male , Neuronal Plasticity/drug effects , Piperazines/administration & dosage , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Sulfides/administration & dosage , Time Factors , Vortioxetine
6.
Neuropharmacology ; 73: 147-59, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23721744

ABSTRACT

Vortioxetine (Lu AA21004) is an investigational novel antidepressant with multimodal activity that functions as a 5-HT3, 5-HT7 and 5-HT(1D) receptor antagonist, 5-HT(1B) receptor partial agonist, 5-HT(1A) receptor agonist and inhibitor of the 5-HT transporter in vitro. Here we explore its anxiolytic and antidepressant potential in adult mice. Vortioxetine was assessed in BalB/cJ@RJ mice using the open-field and forced-swim tests (acute: p.o. 1 h, repeated: daily p.o. 21 days), and in 129S6/SvEvTac mice using the novelty suppressed feeding paradigm (acute: p.o. 1 h, sustained: daily p.o. 14 or 21 days). Fluoxetine and diazepam were controls. Acute and repeated dosing of vortioxetine produced more pronounced anxiolytic- and antidepressant-like activities than fluoxetine. Vortioxetine significantly increased cell proliferation and cell survival and stimulated maturation of immature granule cells in the subgranular zone of the dentate gyrus of the hippocampus after 21 days of treatment. After 14 days, a high dose of vortioxetine increased dendritic length and the number of dendrite intersections, suggesting that vortioxetine accelerates the maturation of immature neurons. Vortioxetine displays an antidepressant and anxiolytic profile following repeated administration associated with increased neurogenesis at several stages. Vortioxetine effects were observed at low levels of 5-HT transporter occupancy, suggesting an alternative mechanism of action to 5-HT reuptake inhibition.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Neurogenesis/drug effects , Piperazines/pharmacology , Sulfides/pharmacology , Animals , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Diazepam/pharmacology , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Fluoxetine/pharmacology , Immobility Response, Tonic/drug effects , Male , Mice , Motor Activity/drug effects , Vortioxetine
SELECTION OF CITATIONS
SEARCH DETAIL