Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Brain Res ; 1789: 147948, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35597327

ABSTRACT

Given the important role of brain-derived neurotrophic factor (BDNF)-mediated Trkß signalling in the mechanism of action of antidepressants (ADs), we examined ligand-receptor interactions in the rat cingulate cortex using a proximity ligation assay (PLA) in response to acute and repeated administration of imipramine (IMI), followed by various drug-free periods. Both the acute and chronic administration of IMI increased the BDNF-Trkß interaction observed 3 h after drug administration. Withdrawal of IMI for 72 h or 7 days did not alter BDNF-Trkß interaction. A significant reduction in this interaction after chronic IMI administration followed by 21 drug-free days was observed, but it returned to the control value when a new dose of IMI was given after this time. The level of mRNA encoding BDNF or Trkß did not change in the experimental groups of animals, so one can conclude that alterations in the BDNF-Trkß interaction depend not on acute vs. repeated treatment with IMI but on the presence of the drug in the body. This effect correlates well with the strong pro-cognitive effect of acute IMI, assessed by the novel object recognition (NOR) test.


Subject(s)
Brain-Derived Neurotrophic Factor , Cognition , Imipramine , Receptor, trkB , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cognition/drug effects , Imipramine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, trkB/genetics , Receptor, trkB/metabolism
2.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34830086

ABSTRACT

BACKGROUND: Cognitive disorders associated with schizophrenia are closely linked to prefrontal cortex (PFC) dysfunction. Administration of the non-competitive NMDA receptor antagonist ketamine (KET) induces cognitive impairment in animals, producing effects similar to those observed in schizophrenic patients. In a previous study, we showed that KET (20 mg/kg) induces cognitive deficits in mice and that administration of clozapine (CLZ) reverses this effect. To identify biochemical mechanisms related to CLZ actions in the context of KET-induced impairment, we performed a biochemical analysis using the same experimental paradigm-acute and sub-chronic administration of these drugs (0.3 and 1 mg/kg). METHODS: Since the effect of CLZ mainly depends on G-protein-related receptors, we used the Signaling PathwayFinder Kit to identify 84 genes involved in GPCR-related signal transduction and then verified the genes that were statistically significantly different on a larger group of mice using RT-PCR and Western blot analyses after the administration of acute and sub-chronic drugs. RESULTS: Of the 84 genes involved in GPCR-related signal transduction, the expression of six, ßarrestin1, ßarrestin2, galanin receptor 2 (GalR2), dopamine receptor 2 (DRD2), metabotropic glutamate receptor 1 (mGluR1), and metabotropic glutamate receptor 5 (mGluR5), was significantly altered. Since these genes affect the levels of other signaling proteins, e.g., extracellular signal-regulated kinase 1/2 (ERK1/2), G protein-coupled receptor kinase 2 (Grk2), and G protein-gated inwardly rectifying potassium 3 (Girk3), we determined their levels in PFC using Western blot. Most of the observed changes occurred after acute treatment with 0.3 mg/kg CLZ. We showed that acute treatment with CLZ at a lower dose significantly increased ßarrestin1 and ERK1/2. KET treatment induced the upregulation of ßarrestin1. Joint administration of these drugs had no effect on the ßarrestin1 level. CONCLUSION: The screening kit we used to study the expression of GPCR-related signal transduction allowed us to select several important genes affected by CLZ. However, the obtained data do not explain the mechanism of action of CLZ that is responsible for reversing KET-induced cognitive impairment.


Subject(s)
Clozapine/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Ketamine/adverse effects , Receptors, G-Protein-Coupled , Animals , Biomarkers/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Ketamine/pharmacology , Male , Mice , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics
3.
Int J Mol Sci ; 21(24)2020 Dec 12.
Article in English | MEDLINE | ID: mdl-33322800

ABSTRACT

In the present study, we aim to identify the effect of restrain stress (RS) on the expression of miRNAs in mouse serum. We used three genotypes of animals (mice with knock-out of the gene-encoding norepinephrine transporter, NET-KO; C57BL/6J, and SWR/J) which had previously been shown to display different sensitivity to RS, and focused on miRNAs which were altered by RS in the serum of all three genotypes. An analysis of miRNAs expression allowed for the identification of a set of 25 differentially expressed miRNAs; 10 were down-regulated compared to an appropriate control group of animals, while 15 were up-regulated. The application of DIANA-miRPath v. 3.0 allowed for the identification of selected pathways (KEGG) and Gene Ontology (GO) categories that were significantly controlled by these miRNAs, while miRWalk v. 3.0-the platform that used the machine learning based algorithm, TaRPmiR-was used to find their targets. The results indicate that 25 miRNAs, identified as altered upon RS in three genotypes of mice, are responsible for regulation of mRNA-encoding proteins that are key for the main hypotheses of depression; therefore, they may help to understand the link between stress and depression at the molecular level.


Subject(s)
Depression/metabolism , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , MicroRNAs/blood , Stress, Physiological/genetics , Algorithms , Animals , Depression/genetics , Down-Regulation , Gene Expression Profiling , Gene Ontology , Machine Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Restraint, Physical/physiology , Signal Transduction/genetics , Up-Regulation
4.
Cells ; 9(4)2020 04 09.
Article in English | MEDLINE | ID: mdl-32283635

ABSTRACT

In the present study, we used three strains of mice with various susceptibility to stress: mice with knock-out of the gene encoding norepinephrine transporter (NET-KO), which are well characterized as displaying a stress-resistant phenotype, as well as two strains of mice displaying two different stress-coping strategies, i.e., C57BL/6J (WT in the present study) and SWR/J. The procedure of restraint stress (RS, 4 h) was applied, and the following behavioral experiments (the forced swim test and sucrose preference test) indicated that NET-KO and SWR/J mice were less sensitive to RS than WT mice. Then, we aimed to find the miRNAs which changed in similar ways in the serum of NET-KO and SWR/J mice subjected to RS, being at the same time different from the miRNAs found in the serum of WT mice. Using Custom TaqMan Array MicroRNA Cards, with primers for majority of miRNAs expressed in the serum (based on a preliminary experiment using the TaqMan Array Rodent MicroRNA A + B Cards Set v3.0, Thermo Fisher Scientific, Waltham, MA, USA) allowed the identification of 21 such miRNAs. Our further analysis focused on miR-1 and miR-155 and their targets-these two miRNAs are involved in the regulation of BDNF expression and can be regarded as biomarkers of stress-resilience.


Subject(s)
MicroRNAs/blood , Norepinephrine Plasma Membrane Transport Proteins/blood , Stress, Physiological/physiology , Animals , Biomarkers/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Prog Mol Biol Transl Sci ; 169: 279-296, 2020.
Article in English | MEDLINE | ID: mdl-31952689

ABSTRACT

Human dopamine D2 receptor (D2R) gene has polymorphic variants, three of them alter its amino acid sequence: Val96Ala, Pro310Ser and Ser311Cys. Their functional role never became the object of extensive studies, even though there are some evidence that they correlate with schizophrenia. The present work reviews data indicating that these mutations play a role in dimer formation with dopamine D1 receptor (D1R), with the strongest effect observed for Ser311Cys variant. Similarly, the affinity for antipsychotic drugs of this genetic variant depends on whether it is expressed together with D1R or not. Better understanding of altered ability of genetic variants of D2R to form dimers with D1R, as well as of altered affinity for antipsychotic drugs, depending on the absence or presence of the second dopamine receptor is of great importance-since these two receptors are not always co-expressed in the same cell. It may well be that targeting new compounds toward the D1R-D2R dimers, which the most probably form under conditions of excessive dopamine release, will result in antipsychotic drugs devoid of serious side effects.


Subject(s)
Antipsychotic Agents/pharmacology , Genetic Variation , Mutation , Receptors, Dopamine/genetics , Alleles , Binding, Competitive , HEK293 Cells , Humans , Ligands , Mutation, Missense , Polymorphism, Single Nucleotide , Protein Multimerization , Receptors, Dopamine/chemistry , Schizophrenia/drug therapy , Schizophrenia/genetics
6.
Neuroscience ; 423: 66-75, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31705885

ABSTRACT

Stress, a major precipitant of depression, and antidepressants have major impact on synaptic integrity and plasticity in brain areas, such as hippocampus (HPC) and prefrontal cortex (PFC). We have recently shown that, unlike Wistar rats, rats of the Wistar-Kyoto (WKY) strain fail to respond to chronic antidepressant treatment after exposure to chronic mild stress (CMS) procedure. However, deep brain stimulation (DBS) of PFC was effective in both strains. We aimed to identify genes that were affected by CMS, to determine whether their expression was normalized by DBS, and to establish whether common changes could be identified in antidepressant responsive (Wistar) and antidepressant-resistant (WKY) strains. Male Wistar and WKY rats were exposed chronically to CMS then treated acutely with DBS. A battery of behavioural tests was used to monitor recovery, followed by TaqMan screening of a panel of genes known to be involved in stress and antidepressant action. WKY showed over-expression of five genes in dorsal HPC and under-expression of seven genes in ventral HPC. Expression of three genes, Egr1, Htr7 and Mmp9 was decreased by CMS and normalized by DBS in the ventral HPC of Wistar rats. Some other changes in gene expression were identified in dorsal HPC and PFC, particularly in Wistars, that were not normalized by DBS. No effects were identified that were common to both Wistars and WKY. The difference between Wistars and WKY in the balance of overall gene expression in HPC may be relevant to the resistance of WKY rats to antidepressant drug treatment.


Subject(s)
Depression/genetics , Stress, Psychological/genetics , Animals , Antidepressive Agents/pharmacology , Behavior Rating Scale , Deep Brain Stimulation , Disease Models, Animal , Genomics , Hippocampus/metabolism , Male , Prefrontal Cortex/metabolism , Rats , Rats, Inbred WKY , Rats, Wistar
7.
Neurosci Lett ; 707: 134292, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31132389

ABSTRACT

The mechanisms underlying the beneficial effects of clozapine (CLZ) in the treatment of schizophrenia still remains far from clear. In the present work we studied the effect of CLZ on the dopamine D2 receptors (D2R) in the mouse brain. CLZ was administered after ketamine (KET) in a paradigm strictly matching the one used in KET-induced attentional set-shifting task (ASST). It has been shown previously that CLZ reversed KET-induced cognitive impairments. In the present study we used in situ hybridization to estimate the level of mRNA, together with specific D2R radioligand, [3H]domperidone binding in the ventral tegmental area (VTA) as well as in the striatum, and observed an increase in the [3H]domperidone binding in the striatum and an increase in D2R mRNA level in the VTA following repeated (but not acute) CLZ administration in mice pre-treated repeatedly with KET. The obtained results allow for conclusion that CLZ in this experimental paradigm enhances biosynthesis of presynaptic D2R.


Subject(s)
Antipsychotic Agents/pharmacology , Brain/drug effects , Clozapine/pharmacology , Cognition Disorders/metabolism , Dopamine D2 Receptor Antagonists , Dopamine/metabolism , Ketamine , Receptors, Dopamine D2/metabolism , Animals , Antipsychotic Agents/administration & dosage , Brain/metabolism , Clozapine/administration & dosage , Cognition Disorders/chemically induced , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism , Receptors, Dopamine D2/genetics , Schizophrenic Psychology , Signal Transduction
8.
Methods Cell Biol ; 149: 155-178, 2019.
Article in English | MEDLINE | ID: mdl-30616817

ABSTRACT

Initially G protein-coupled receptors, GPCRs, were thought to act as monomers, but recently strong evidence has been gathered indicating that they are capable of forming homo- and heterodimers or higher order oligomeric complexes, and that the dimerization phenomenon can modulate the pharmacological response and function of these receptors. In this chapter we point to the great potential of alternative therapeutic approach targeted at GPCR dimers, which is especially important in the field of neuropsychopharmacology. We also included a brief description of methods used for studying the phenomenon of GPCR oligomerization, with particular attention paid to the proximity ligation assay, PLA, the procedure which allows the study of interactions between receptors not only in vitro but also in vivo, with good anatomical resolution, what is especially important in the studies of various GPCRs involved in central neurotransmission.


Subject(s)
Biological Assay/methods , Protein Multimerization , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line, Tumor , Humans , Rats
10.
Psychopharmacology (Berl) ; 235(7): 2137-2149, 2018 07.
Article in English | MEDLINE | ID: mdl-29713785

ABSTRACT

RATIONALE: The role of somatostatin and its receptors for the stress-related neuropsychiatric disorders has been widely raised. Recently, we have also demonstrated the involvement of somatostatin receptor type 2-sst2R and dopamine receptor type 2-D2R in stress. OBJECTIVE: In this context, we decided to find if these receptors are involved in response to antidepressant treatment in animal model of depression-chronic mild stress (CMS). METHODS: Here, we report data obtained following 7-week CMS procedure. The specific binding of [125I]Tyr3-Octreotide to sst2R and [3H]Domperidone to D2R was measured in the rat brain, using autoradiography. Additionally, the level of dopamine and metabolites was measured in the rat brain. RESULTS: In the final baseline test after 7 weeks of stress, the reduced consumption of sucrose solution was observed (controls vs the stressed animals (6.25 0.16 vs. 10.39 0.41; p < 0.05). Imipramine was administered for the next 5 weeks, and it reversed anhedonia in majority of animals (imipramine-reactive); however, in some animals, it did not (imipramine-non-reactive). Two-way repeated measures ANOVA revealed significant effects of stress and treatment and time interaction [F(16, 168) = 3.72; p < 0.0001], n = 10 per groups. We observed decreased binding of [125I]Tyr3-Octreotide in most of rat brain regions in imipramine non-reactive groups of animals. The decrease of D2R after stress in striatum and nucleus accumbens and no effect of imipramine were observed. In the striatum and prefrontal cortex, the significant role of stress and imipramine in dopamine levels was observed. CONCLUSIONS: The results obtained in binding assays, together with dopamine level, indicate the involvement of sst2R receptors for reaction to antidepressant treatment. Besides, the stress context itself changes the effect of antidepressant drug.


Subject(s)
Brain/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Somatostatin/metabolism , Stress, Psychological/metabolism , Anhedonia/drug effects , Animals , Antidepressive Agents/pharmacology , Autoradiography , Brain/diagnostic imaging , Brain/drug effects , Depression/diagnostic imaging , Depression/metabolism , Disease Models, Animal , Domperidone/metabolism , Dopamine/metabolism , Imipramine/pharmacology , Iodine Radioisotopes , Male , Neostriatum/drug effects , Neostriatum/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Octreotide/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Receptors, Dopamine D2/drug effects , Receptors, Somatostatin/drug effects , Stress, Psychological/diagnostic imaging , Sucrose , Tritium
11.
Behav Pharmacol ; 29(6): 537-542, 2018 09.
Article in English | MEDLINE | ID: mdl-29537988

ABSTRACT

We have previously reported the effects of intracranial injections of dopamine D1, D2 and D3 ligands in animals subjected to the Novel Object Recognition (NOR) test following exposure to chronic mild stress (CMS) and chronic treatment with risperidone (RSP). Here, we present some molecular biological data from the same animals. It was predicted that brain-derived neurotrophic factor (BDNF) signalling in the prefrontal cortex (PFC) would reflect behavioural performance, implying an increase following acute administration of a D2 agonist or a D3 antagonist, blockade of this effect by CMS and its restoration by chronic RSP. In separate cohorts, animals were injected within the PFC or the hippocampus (HPC) with either the D1 agonist SKF-81297, the D2 agonist quinpirole or the D3 antagonist SB-277,011, following exposure to control conditions or CMS and chronic treatment with saline or RSP. Intracranial injections followed an exposure trial in the NOR test, with a retention trial 24 h later. Immediately afterwards, the animals were killed and expression of BDNF and TRKß protein, and their respective mRNAs, was measured in PFC and HPC samples. CMS decreased the expression of TRKß in both PFC and HPC. Several effects associated with intracranial injection were noted, but they were inconsistent and unrelated to CMS exposure. The effects of CMS on TRKß are consistent with a decrease in BDNF signalling, albeit that expression of BDNF itself did not change significantly. There was no evidence for an involvement of the BDNF-TRKß system in responses to RSP or dopamine ligands in animals exposed to CMS. However, there was a 24 h delay between the intracranial injection and tissue harvesting, meaning that brief early drug effects could have been missed.


Subject(s)
Antipsychotic Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Receptors, Dopamine/metabolism , Risperidone/therapeutic use , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Animals , Benzazepines/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Dopamine Agents/pharmacology , Electroencephalography , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Quinpirole/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar
12.
Neuroscience ; 377: 184-196, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29544901

ABSTRACT

Recently, it has been shown that serotonin 5-HT1A receptor interacts with dopamine D2 receptor in vitro. However, the existence of 5-HT1A-D2 heteromers in native tissue remains unexplored. In the present study, we investigated 5-HT1A-D2 receptor heteromerization in mice treated acutely or chronically with paroxetine (10 mg/kg) or risperidone (0.05 mg/kg). Receptor heteromerization was visualized and quantified in the mouse brain by in situ proximity ligation assay (PLA). Additionally, we aimed to determine the cellular localization of 5-HT1A-D2 receptor heteromers in mouse adult primary neuronal cells by immunofluorescent staining with markers for astrocytes (GFAP) and neurons (NeuN and MAP2). The results from the current study demonstrated that 5-HT1A and D2 receptor co-localization and heteromerization occurred in the mouse prefrontal cortex. Counterstaining after PLA confirmed neuronal (pyramidal and GABAergic) as well as astrocytal localization of 5-HT1A-D2 receptor heteromers. Chronic administration of paroxetine or risperidone increased the level of 5-HT1A-D2 receptor heteromers in the prefrontal cortex. These changes were not accompanied by any changes in the expression of mRNAs (measured by in situ hybridization) or densities of 5-HT1A and D2 receptors (quantified by receptor autoradiography with [3H]8-OH-DPAT and [3H]domperidone, respectively), what all indicated that paroxetine and risperidone facilitated 5-HT1A-D2 heteromer formation independently of the receptor expression. In vitro homogenous time-resolved FRET (HTRF) study confirmed the ability of tested drugs to influence the human 5-HT1A-D2 heteromer formation. The obtained data indicate that the increase in 5-HT1A-D2 receptor heteromerization is a common molecular characteristic of paroxetine and low-dose risperidone treatment.


Subject(s)
Neurotransmitter Agents/pharmacology , Paroxetine/pharmacology , Prefrontal Cortex/drug effects , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Dopamine D2/metabolism , Risperidone/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cricetulus , Humans , Male , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Protein Multimerization/drug effects , RNA, Messenger/metabolism
13.
Front Mol Neurosci ; 11: 40, 2018.
Article in English | MEDLINE | ID: mdl-29497362

ABSTRACT

G-protein-coupled receptor (GPCR) heterodimers are new targets for the treatment of schizophrenia. Dopamine D2 receptors and serotonin 5-HT1A and 5-HT2A receptors play an important role in neurotransmission and have been implicated in many human psychiatric disorders, including schizophrenia. Therefore, in this study, we investigated whether antipsychotic drugs (clozapine (CLZ) and haloperidol (HAL)) affected the formation of heterodimers of D2-5-HT1A receptors as well as 5-HT1A-5-HT2A receptors. Proximity ligation assay (PLA) was used to accurately visualize, for the first time, GPCR heterodimers both at in vitro and ex vivo levels. In line with our previous behavioral studies, we used ketamine to induce cognitive deficits in mice. Our study confirmed the co-localization of D2/5-HT1A and 5-HT1A/5-HT2A receptors in the mouse cortex. Low-dose CLZ (0.3 mg/kg) administered repeatedly, but not CLZ at 1 mg/kg, increased the level of D2-5-HT1A and 5-HT1A-5-HT2A heterodimers in the mouse prefrontal and frontal cortex. On the other hand, HAL decreased the level of GPCR heterodimers. Ketamine affected the formation of 5-HT1A-5-HT2A, but not D2-5-HT1A, heterodimers.

14.
Brain Res Bull ; 135: 92-97, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28987282

ABSTRACT

The interaction between the dopaminergic and somatostatinergic systems is considered to play a potential role in mood regulation. Chronic administration of antidepressants influences release of both neurotransmitters. The molecular basis of the functional cooperation may stem from the physical interaction of somatostatin receptor subtypes and dopamine D2 receptors since they colocalize in striatal interneurons and were shown to undergo ligand-dependent heterodimerization in heterologous expression systems. In present study we adapted in situ proximity ligation assay to investigate the occurrence of D2-Sst5 receptor heterocomplexes, and their possible alterations in the striatum of mice treated acutely and repeatedly (21days) with antidepressant drugs of different pharmacological profiles (escitalopram and desipramine). Additionally we analysed number of heterocomplexes in primary striatal neuronal cultures incubated with both antidepressant drugs for 1h and 6days. The studies revealed that antidepressants increase formation of D2-Sst5 receptors heterodimers. These findings provide interesting evidence that dopamine D2 and somatostatin Sst5 heterodimers may be considered as potential mediators of antidepressant effects, since the heterodimerization of these receptors occurs in native brain tissue as well as in primary striatal neuronal cultures where receptors are expressed at physiological levels.


Subject(s)
Corpus Striatum/drug effects , Receptors, Somatostatin/drug effects , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Cell Culture Techniques , Corpus Striatum/metabolism , Dopamine/metabolism , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Multiprotein Complexes/drug effects , Neostriatum/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism
15.
Mol Neurobiol ; 54(8): 5741-5751, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27660265

ABSTRACT

Prolonged stress perturbs physiological balance of a subject and thus can lead to depression. Nevertheless, some individuals are more resilient to stress than the others. Defining molecular factors underlying resilience to stress may contribute to the development of a new antidepressant strategy based on the restoration of resilient phenotype in depressed subjects. We used chronic mild stress (CMS) paradigm-well-characterized animal model of depression which caused in rats behavioral deficits (anhedonia) manifested by decreased consumption of sucrose solution. CMS also generated a proportion of resilient rats which did not alter sucrose consumption despite being stressed. Recently, regulation of a gene expression associated with microRNA (miRNA) is considered as an important factor modulating biochemical response to stress. Based on our previous work and literature survey, we investigated changes in the expression level of seven miRNAs (i.e., miR-18a-5p, miR-34a-5p, miR-135a-5p, miR-195-5p, miR-320-3p, miR-674-3p, miR-872-5p) in mesocortical circuit-crucially involved in stress response in order to find differences between susceptible and resilient phenotype. Bioinformatic analysis showed that all miRNAs of interest potentially target serotonin transporter (SERT). Chronic stress caused global increase in the expression of the abovementioned miRNAs in ventral tegmental area (VTA) of stressed rats followed by parallel decrease in miRNA expression in prefrontal cortex (PCx). This effect was more profound in resilient than anhedonic animals. Moreover, we observed decreased level of SERT in VTA of resilient rats. Our findings show that mesocortical circuit is involved in the response to stress and this phenomenon is more efficient in resilient animals.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Gene Expression/drug effects , MicroRNAs/metabolism , Stress, Psychological/metabolism , Anhedonia/drug effects , Animals , Depression/genetics , Disease Models, Animal , Male , Prefrontal Cortex/metabolism , Rats, Wistar , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Stress, Psychological/genetics
16.
Eur Neuropsychopharmacol ; 26(1): 23-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26628105

ABSTRACT

MicroRNAs (miRNAs) are involved in stress-related pathologies. However, the molecular mechanisms underlying stress resilience are elusive. Using chronic mild stress (CMS), an animal model of depression, we identified animals exhibiting a resilient phenotype. We investigated serum levels of corticosterone, melatonin and 376 mature miRNAs to find peripheral biomarkers associated with the resilient phenotype. miR-16, selected during screening step, was assayed in different brain regions in order to find potential relationship between brain and peripheral alterations in response to stress. Two CMS experiments that lasted for 2 and 7 consecutive weeks were performed. During both CMS procedures, sucrose consumption levels were significantly decreased in anhedonic-like animals (p<0.0001) compared with unstressed animals, whereas the drinking profiles of resilient rats did not change despite the rats being stressed. Serum corticosterone measurements indicated that anhedonic-like animals had blunted hypothalamic-pituitary-adrenal (HPA) axis activity, whereas resilient animals exhibited dynamic responses to stress. miRNA profiling revealed that resilient animals had elevated serum levels of miR-16 after 7 weeks of CMS (adjusted p-value<0.007). Moreover, resilient animals exhibited reciprocal changes in miR-16 expression level in mesocortical pathway after 2 weeks of CMS (p<0.008). A bioinformatic analysis showed that miR-16 regulates genes involved in the functioning of the nervous system in both humans and rodents. Resilient animals can actively cope with stress on a biochemical level and miR-16 may contribute to a "stress-resistant" behavioral phenotype by pleiotropic modulation of the expression of genes involved in the function of the nervous system.


Subject(s)
Depressive Disorder/metabolism , Limbic System/metabolism , MicroRNAs/metabolism , Resilience, Psychological , Stress, Psychological/metabolism , Anhedonia/physiology , Animals , Biomarkers/metabolism , Chronic Disease , Corticosterone/blood , Dietary Sucrose , Disease Models, Animal , Feeding Behavior , Male , Melatonin/blood , Random Allocation , Rats, Wistar , Time
17.
J Neurochem ; 134(4): 704-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031442

ABSTRACT

We have previously demonstrated that nicotine withdrawal produces depression-like behavior and that serotonin (5-HT)2A/2C receptor ligands modulate that mood-like state. In the present study we aimed to identify the mechanisms (changes in radioligand binding, transcription or RNA-editing) related to such a behavioral outcome. Rats received vehicle or nicotine (0.4 mg/kg, s.c.) for 5 days in home cages. Brain 5-HT2A/2C receptors were analyzed on day 3 of nicotine withdrawal. Nicotine withdrawal increased [(3)H]ketanserin binding to 5-HT2A receptors in the ventral tegmental area and ventral dentate gyrus, yet decreased binding in the nucleus accumbens shell. Reduction in [(3)H]mesulergine binding to 5-HT2C receptors was seen in the ventral dentate gyrus. Profound decrease in the 5-HT2A receptor transcript level was noted in the hippocampus and ventral tegmental area. Out of five 5-HT2C receptor mRNA editing sites, deep sequencing data showed a reduction in editing at the E site and a trend toward reduction at the C site in the hippocampus. In the ventral tegmental area, a reduction for the frequency of CD 5-HT2C receptor transcript was seen. These results show that the reduction in the 5-HT2A receptor transcript level may be an auto-regulatory response to the increased receptor density in the hippocampus and ventral tegmental area during nicotine withdrawal, while decreased 5-HT2C receptor mRNA editing may explain the reduction in receptor labeling in the hippocampus. Serotonin (5-HT)2A/2C receptor ligands alleviate depression-like state in nicotine-withdrawn rats. Here, we show that the reduction in 5-HT2A receptor transcript level may be an auto-regulatory response to the increased receptor number in the hippocampus and ventral tegmental area during nicotine withdrawal, while attenuated 5-HT2C receptor mRNA editing in the hippocampus might explain reduced inverse agonist binding to 5-HT2C receptor and suggest a shift toward a population of more active receptors. 5-HT, serotonin; 5-HT2A R, 5-HT2A receptor; 5-HT2C R, 5-HT2C receptor.


Subject(s)
Brain/metabolism , Nicotine/adverse effects , Receptor, Serotonin, 5-HT2A/physiology , Receptor, Serotonin, 5-HT2C/physiology , Substance Withdrawal Syndrome/metabolism , Animals , Brain/drug effects , Immobilization/psychology , Male , Nicotine/administration & dosage , Rats , Rats, Wistar , Substance Withdrawal Syndrome/psychology
18.
Eur Neuropsychopharmacol ; 25(8): 1099-108, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26002194

ABSTRACT

These studies aimed to identify the genes differentially expressed in the frontal cortex of mice bearing a life-long norepinephrine transporter knock-out (NET-KO) and wild-type animals (WT). Differences in gene expression in the mouse frontal cortex were studied using a whole-genome microarray approach. Using an alternative approach, i.e. RT-PCR (reverse transcription polymerase chain reaction) with primers complementary to various exons of the NET gene, as well as TaqMan arrays, the level of mRNA encoding the NET in other brain regions of the NET-KO mice was also examined. The analyses revealed a group of 92 transcripts (27 genes) that differentiated the NET-KO mice from the WT mice. Surprisingly, the studies have shown that the mRNA encoding NET accumulated in the brain regions rich in norepinephrine nerve endings in the NET-KO mice. Because there is no other source of NET mRNA besides the noradrenergic terminals in the brain regions studied, these results might speak in favor of the presence of mRNA in axon terminals. RNA-Binding Protein Immunoprecipitation approach indicated that mRNA encoding NET was detected in the Ago2 protein/mRNA complex. In addition, the amount of Ago2 protein in the frontal cortex was significantly higher in NET-KO mice as compared with that of the WT animals. These results are important for further characterization of the NET-KO mice, which - besides other merits - might serve as a good model to study the fate of truncated mRNA in neurons.


Subject(s)
Brain/metabolism , Neurons/metabolism , Norepinephrine Plasma Membrane Transport Proteins/deficiency , Norepinephrine/metabolism , Animals , Argonaute Proteins/metabolism , Blotting, Western , Gene Expression , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Norepinephrine Plasma Membrane Transport Proteins/genetics , Polymerase Chain Reaction , RNA, Messenger/metabolism
19.
Pharmacol Rep ; 67(2): 345-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712661

ABSTRACT

BACKGROUND: These studies aimed to identify the genes differentially expressed in the frontal cortex of mice treated repeatedly with either saline or desipramine (DMI). METHODS: Differences in gene expression in the mouse frontal cortex were studied using a whole-genome microarray approach. RESULTS: The analyses revealed a group of 88 transcripts (18 genes) that were differentially expressed between the mice treated with saline and those treated with DMI. These genes include Spnb2, Mef2c, Ncam1, Hsp90ab1, Kif1b, Ddx6 and Gsk3b, which were connected in the gene relationship network. CONCLUSIONS: It appears that one week of DMI administration measurably altered the expression of a small number of genes, including genes connected with neuroplasticity and cytoskeletal changes, the regulation of calcium levels in the cell or translation processes.


Subject(s)
Desipramine/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Gene Expression Profiling , Gene Expression/drug effects , Oligonucleotide Array Sequence Analysis , Animals , Male , Mice
20.
Brain Res ; 1594: 284-92, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25451113

ABSTRACT

Norepinephrine transporter knock-out mice (NET-KO) exhibit depression-resistant phenotypes. They manifest significantly shorter immobility times in both the forced swim test and the tail suspension test. Moreover, biochemical studies have revealed the up-regulation of other monoamine transporters (dopamine and serotonin) in the brains of NET-KO mice, similar to the phenomenon observed after the chronic pharmacological blockade of norepinephrine transporter by desipramine in wild-type (WT) animals. NET-KO mice are also resistant to stress, as we demonstrated previously by measuring plasma corticosterone concentration. In the present study, we used a microdissection technique to separate target brain regions and the TaqMan Low Density Array approach to test the expression of a group of genes in the NET-KO mice compared with WT animals. A group of genes with altered expression were identified in four brain structures (frontal and cingulate cortices, dentate gyrus of hippocampus and basal-lateral amygdala) of NET-KO mice compared with WT mice. These genes are known to be altered by antidepressant drugs administration. The most interesting gene is Crh-bp, which modulates the activity of corticotrophin--releasing hormone (CRH) and several CRH-family members. Generally, genetic disturbances within noradrenergic neurons result in biological changes, such as in signal transduction and intercellular communication, and may be linked to changes in noradrenaline levels in the brains of NET-KO mice.


Subject(s)
Brain , Nerve Growth Factors/biosynthesis , Neuropeptides/biosynthesis , Norepinephrine Plasma Membrane Transport Proteins/genetics , Transcriptome , Animals , Antidepressive Agents/pharmacology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdissection , Nerve Growth Factors/genetics , Neuropeptides/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...