Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int J Neuropsychopharmacol ; 27(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38457375

RESUMO

BACKGROUND: Major depressive disorder (MDD) is a leading cause of disability with significant mortality risk. Despite progress in our understanding of the etiology of MDD, the underlying molecular changes in the brain remain poorly understood. Extracellular vesicles (EVs) are lipid-bound particles that can reflect the molecular signatures of the tissue of origin. We aimed to optimize a streamlined EV isolation protocol from postmortem brain tissue and determine whether EV RNA cargo, particularly microRNAs (miRNAs), have an MDD-specific profile. METHODS: EVs were isolated from postmortem human brain tissue. Quality was assessed using western blots, transmission electron microscopy, and microfluidic resistive pulse sensing. EV RNA was extracted and sequenced on Illumina platforms. Functional follow-up was performed in silico. RESULTS: Quality assessment showed an enrichment of EV markers, as well as a size distribution of 30 to 200 nm in diameter, and no contamination with cellular debris. Small RNA profiling indicated the presence of several RNA biotypes, with miRNAs and transfer RNAs being the most prominent. Exploring miRNA levels between groups revealed decreased expression of miR-92a-3p and miR-129-5p, which was validated by qPCR and was specific to EVs and not seen in bulk tissue. Finally, in silico functional analyses indicate potential roles for these 2 miRNAs in neurotransmission and synaptic plasticity. CONCLUSION: We provide a streamlined isolation protocol that yields EVs of high quality that are suitable for molecular follow-up. Our findings warrant future investigations into brain EV miRNA dysregulation in MDD.


Assuntos
Transtorno Depressivo Maior , Vesículas Extracelulares , MicroRNAs , Humanos , Transtorno Depressivo Maior/metabolismo , Depressão , MicroRNAs/genética , Vesículas Extracelulares/genética , Encéfalo/metabolismo
2.
Neuron ; 111(2): 144-146, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36657396

RESUMO

In this issue of Neuron, Namkung et al. demonstrate that the microRNA miR-124-3p is associated with polygenic risk scores shared between schizophrenia and bipolar disorder and confers risk to behavioral alterations common to these two disorders through modulation of AMPA receptor neurotransmission.


Assuntos
Transtorno Bipolar , MicroRNAs , Esquizofrenia , Humanos , Transtorno Bipolar/genética , Esquizofrenia/genética , MicroRNAs/genética , Herança Multifatorial/genética , Fatores de Risco , Predisposição Genética para Doença/genética
3.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34768740

RESUMO

Depression is an effect of complex interactions between genetic, epigenetic and environmental factors. It is well established that stress responses are associated with multiple modest and often dynamic molecular changes in the homeostatic balance, rather than with a single genetic factor that has a strong phenotypic penetration. As depression is a multifaceted phenotype, it is important to study biochemical pathways that can regulate the overall allostasis of the brain. One such biological system that has the potential to fine-tune a multitude of diverse molecular processes is RNA interference (RNAi). RNAi is an epigenetic process showing a very low level of evolutionary diversity, and relies on the posttranscriptional regulation of gene expression using, in the case of mammals, primarily short (17-23 nucleotides) noncoding RNA transcripts called microRNAs (miRNA). In this review, our objective was to examine, summarize and discuss recent advances in the field of biomedical and clinical research on the role of miRNA-mediated regulation of gene expression in the development of depression. We focused on studies investigating post-mortem brain tissue of individuals with depression, as well as research aiming to elucidate the biomarker potential of miRNAs in depression and antidepressant response.


Assuntos
Depressão/genética , Transtorno Depressivo Maior/genética , MicroRNAs/genética , Animais , Evolução Biológica , Epigênese Genética/genética , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Humanos , Herança Multifatorial/genética , Interferência de RNA/fisiologia , Transcriptoma/genética
4.
FEBS J ; 288(22): 6563-6580, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34058074

RESUMO

Monocyte chemoattractant protein-induced protein 1 (MCPIP1, alias Regnase 1) is a negative regulator of inflammation, acting through cleavage of transcripts coding for proinflammatory cytokines and by inhibition of NFκB activity. Moreover, it was demonstrated that MCPIP1 regulates lipid metabolism both in adipose tissue and in hepatocytes. In this study, we investigated the effects of tissue-specific Mcpip1 deletion on the regulation of hepatic metabolism and development of nonalcoholic fatty liver disease (NAFLD). We used control Mcpip1fl/fl mice and animals with deletion of Mcpip1 in myeloid leukocytes (Mcpip1fl/fl LysMCre ) and in hepatocytes (Mcpip1fl/fl AlbCre ), which were fed chow or a high-fat diet (HFD) for 12 weeks. Mcpip1fl/fl LysMCre mice fed a chow diet were characterized by a significantly reduced hepatic expression of genes regulating lipid and glucose metabolism, which subsequently resulted in low plasma glucose level and dyslipidemia. These animals also displayed systemic inflammation, demonstrated by increased concentrations of cytokines in the plasma and high Tnfa, Il6, IL1b mRNA levels in the liver and brown adipose tissue (BAT). Proinflammatory leukocyte infiltration into BAT, together with low expression of Ucp1 and Ppargc1a, resulted in hypothermia of 22-week-old Mcpip1fl/fl LysMCre mice. On the other hand, there were no significant changes in phenotype in Mcpip1fl/fl AlbCre mice. Although we detected a reduced hepatic expression of genes regulating glucose metabolism and ß-oxidation in these mice, they remained asymptomatic. Upon feeding with a HFD, Mcpip1fl/fl LysMCre mice did not develop obesity, glucose intolerance, nor hepatic steatosis, but were characterized by low plasma glucose level and dyslipidemia, along with proinflammatory phenotype. Mcpip1fl/fl AlbCre animals, following a HFD, became hypercholesterolemic, but accumulated lipids in the liver at the same level as Mcpip1fl/fl mice, and no changes in the level of soluble factors tested in the plasma were detected. We have demonstrated that Mcpip1 protein plays an important role in the liver homeostasis. Depletion of Mcpip1 in myeloid leukocytes, followed by systemic inflammation, has a more pronounced effect on controlling liver metabolism and homeostasis than the depletion of Mcpip1 in hepatocytes.


Assuntos
Fígado Gorduroso/metabolismo , Fígado/metabolismo , Células Mieloides/metabolismo , Obesidade/metabolismo , Ribonucleases/metabolismo , Animais , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ribonucleases/sangue , Ribonucleases/deficiência
5.
IUBMB Life ; 72(8): 1807-1818, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32593213

RESUMO

It has been previously established that hypoxia leads to tumor development, treatment resistance, and a poor prognosis. Under oxygen deprivation, hypoxia-inducible factors (HIFs) are stimulated to activate the genes necessary for tumor development in a low-oxygen environment. These genes encode regulators of angiogenesis, epithelial-mesenchymal transition, and cellular metabolism. A disulfide isomerase, anterior gradient 2 (AGR2), has been shown to increase hypoxia-inducible factor 1, alpha subunit (HIF-1α) stability in breast cancer. Our goal was to determine if AGR2 affects the level of transcription factor hypoxia-inducible factor 2, alpha subunit (HIF-2α). As a model, we used the clear cell renal cell carcinoma (ccRCC) cell line Caki-1. The cells were transduced with lentiviral vector (Tet-On) encoding AGR2. After induction of AGR2 expression, cells were grown under either hypoxic (0.5% O2 ) or normoxic (21% O2 ) conditions. Our data showed that AGR2 upregulated both HIF-1α and HIF-2α expression in Caki-1 cells increasing the expression of HIF-activated genes (glucose transporter 1, phosphoglycerate kinase 1, vascular endothelial growth factor A, and transforming growth factor-alpha) under the hypoxic conditions. Under the normoxic conditions, AGR2 strongly activated CCAAT-enhancer binding protein beta (C/EBPß). Upregulation of C/EBPß correlated with increased expression and secretion of the interleukin-6 and interleukin-8, inducing angiogenesis and inflammation in Caki-1 cells. In summary, our studies revealed that AGR2 has essential functions in ccRCC progression through upregulation of C/EBPß and HIF-2α expressions, which affects cell signaling and metabolism.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Carcinoma de Células Renais/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mucoproteínas/genética , Proteínas Oncogênicas/genética , Carcinogênese/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Interleucina-6/genética , Interleucina-8/genética , Proteínas de Neoplasias/genética , Transdução de Sinais/genética , Fator A de Crescimento do Endotélio Vascular/genética
6.
Mol Neurobiol ; 56(11): 7680-7693, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31098953

RESUMO

Disturbed serotonergic signaling in the hippocampus observed in many individuals vulnerable to stress has been suggested as one of the primary factors contributing to the development of depression. However, little is known about the physiology of the brain in the resilient phenotype. Resilient subjects maintain a positive mood and psychological balance despite being under the stress influence. In our study, we generated stress-vulnerable and resilient rats by using a chronic mild stress (CMS) paradigm. Using different molecular approaches, we revealed that resilient animals exhibited a significantly decreased expression level of miR-18a-5p and, in the same time, an elevated level of 5-HT1AR in dorsal, but not ventral, part of the hippocampus. Described biochemical changes were not observed in animals behaviorally vulnerable to stress. Further, in vitro analysis showed that miR-18a-5p may be a negative epigenetic regulator of 5-HT1AR since the treatment of adult hippocampal neurons with miR-18a-5p mimic significantly lowered the expression level of mRNA encoding 5-HT1AR. Moreover, bioinformatic analysis of potential target genes expressed in the hippocampus and being regulated by miR-18a-5p showed that this microRNA may regulate biological processes, such as axonogenesis, which are important in the functioning of the hippocampus in both rats and humans. All these molecular features may contribute to serotonergic homeostatic balance at the level of serotonin turnover observed in hippocampi of resilient but not stress-vulnerable rats. Delineation of further molecular and biochemical markers underlying resilience to stress may contribute to the development of new antidepressant strategies which will restore resilient phenotype in depressed patients.


Assuntos
Hipocampo/metabolismo , MicroRNAs/genética , Receptor 5-HT1A de Serotonina/genética , Resiliência Psicológica , Estresse Psicológico/genética , Animais , Encéfalo/metabolismo , Corticosterona/sangue , Regulação da Expressão Gênica , Ácido Hidroxi-Indolacético/metabolismo , Masculino , MicroRNAs/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Esteroides/sangue , Sacarose
7.
Methods Cell Biol ; 149: 155-178, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30616817

RESUMO

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.


Assuntos
Bioensaio/métodos , Multimerização Proteica , Receptores Acoplados a Proteínas G/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Ratos
8.
Psychopharmacology (Berl) ; 235(7): 2137-2149, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29713785

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Estresse Psicológico/metabolismo , Anedonia/efeitos dos fármacos , Animais , Antidepressivos/farmacologia , Autorradiografia , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Depressão/diagnóstico por imagem , Depressão/metabolismo , Modelos Animais de Doenças , Domperidona/metabolismo , Dopamina/metabolismo , Imipramina/farmacologia , Radioisótopos do Iodo , Masculino , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Octreotida/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Wistar , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Somatostatina/efeitos dos fármacos , Estresse Psicológico/diagnóstico por imagem , Sacarose , Trítio
9.
Front Mol Neurosci ; 11: 40, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29497362

RESUMO

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.

10.
Neuroscience ; 377: 184-196, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29544901

RESUMO

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.


Assuntos
Neurotransmissores/farmacologia , Paroxetina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Dopamina D2/metabolismo , Risperidona/farmacologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , Cricetulus , Humanos , Masculino , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/metabolismo , Multimerização Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo
11.
Schizophr Res ; 192: 423-430, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28433499

RESUMO

It has been shown that the metabotropic glutamate receptor subtype 5 (mGluR5) is functionally associated with the NMDA subtype of the glutamate receptor family (NMDA receptors). These two receptors colocalize in brain regions associated with schizophrenia. Although the role of the NMDA receptor in cognitive and negative symptoms of schizophrenia is well studied, information about the role of mGluR5 receptors in schizophrenia is sparse. In our work, we show that subchronic administration of ketamine, a well-studied, non-competitive antagonist of NMDA receptors, caused cognitive deficits in rats as shown by testing novel object recognition (NOR). Moreover, we reveal that subchronic administration of ketamine increased the mRNA and protein expression levels of mGluR5 receptors in regions CA1 and CA3 of the dorsal part of the hippocampus, both of which are strongly associated with the formation of visual memory, which is tested via NOR. We postulate that increased expression of mGluR5 receptors in the dorsal part of the hippocampus may reflect compensatory changes to imbalanced glutamate neurotransmission associated with the hypoactivation of NMDA receptors.


Assuntos
Receptor de Glutamato Metabotrópico 5/metabolismo , Esquizofrenia/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Região CA3 Hipocampal/metabolismo , Região CA3 Hipocampal/patologia , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Ketamina , Masculino , Parvalbuminas/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Dopamina D2/metabolismo , Reconhecimento Psicológico/fisiologia , Esquizofrenia/patologia
12.
Brain Res Bull ; 135: 92-97, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28987282

RESUMO

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.


Assuntos
Corpo Estriado/efeitos dos fármacos , Receptores de Somatostatina/efeitos dos fármacos , Animais , Antidepressivos/metabolismo , Antidepressivos/farmacologia , Técnicas de Cultura de Células , Corpo Estriado/metabolismo , Dopamina/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/efeitos dos fármacos , Neostriado/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo
13.
Mol Neurobiol ; 54(8): 5741-5751, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27660265

RESUMO

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.


Assuntos
Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Expressão Gênica/efeitos dos fármacos , MicroRNAs/metabolismo , Estresse Psicológico/metabolismo , Anedonia/efeitos dos fármacos , Animais , Depressão/genética , Modelos Animais de Doenças , Masculino , Córtex Pré-Frontal/metabolismo , Ratos Wistar , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Estresse Psicológico/genética
14.
Eur Neuropsychopharmacol ; 26(1): 23-36, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26628105

RESUMO

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.


Assuntos
Transtorno Depressivo/metabolismo , Sistema Límbico/metabolismo , MicroRNAs/metabolismo , Resiliência Psicológica , Estresse Psicológico/metabolismo , Anedonia/fisiologia , Animais , Biomarcadores/metabolismo , Doença Crônica , Corticosterona/sangue , Sacarose Alimentar , Modelos Animais de Doenças , Comportamento Alimentar , Masculino , Melatonina/sangue , Distribuição Aleatória , Ratos Wistar , Tempo
15.
Eur Neuropsychopharmacol ; 25(8): 1099-108, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26002194

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/deficiência , Norepinefrina/metabolismo , Animais , Proteínas Argonautas/metabolismo , Western Blotting , Expressão Gênica , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo
16.
Pharmacol Rep ; 67(2): 345-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25712661

RESUMO

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.


Assuntos
Desipramina/farmacologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Perfilação da Expressão Gênica , Expressão Gênica/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Masculino , Camundongos
17.
Brain Res ; 1594: 284-92, 2015 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-25451113

RESUMO

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.


Assuntos
Encéfalo , Fatores de Crescimento Neural/biossíntese , Neuropeptídeos/biossíntese , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Transcriptoma , Animais , Antidepressivos/farmacologia , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdissecção , Fatores de Crescimento Neural/genética , Neuropeptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Peptides ; 61: 98-106, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25236411

RESUMO

The purpose of this study was to examine molecular markers of the stress response at the pituitary and peripheral levels in animals that responded differently to chronic mild stress (CMS). Rats were subjected to 2-weeks CMS and symptoms of anhedonia was measured by the consumption of 1% sucrose solution. mRNA levels of CRH-family neuropeptides (Crh-corticotropin-releasing hormone, Ucn1-urocortin 1, Ucn2-urocortin 2, Ucn3-urocortin 3), CRH receptors (Crhr1-corticotropin-releasing hormone receptor 1, Crhr2-corticotropin-releasing hormone receptor 2) and Crhbp (corticotropin-releasing factor binding protein) in the pituitaries of rats were determined with real-time PCR. Plasma levels of ACTH (adrenocorticotropin), CRH and urocortins were measured with ELISA assays. CMS procedure led to the development of anhedonia manifested by the decreased sucrose consumption (stress-reactive, SR, stress-susceptible group). Additionally, the group of animals not exhibiting any signs of anhedonia (stress non-reactive, SNR, stress-resilient group) and the group characterized by the increased sucrose consumption (stress invert-reactive group SIR) were selected. The significant increases in ACTH plasma level accompanied by the decreases in the pituitary gene expression of the Crh, Ucn2 and Ucn3 in both stress non-reactive and stress invert-reactive groups were observed. The only molecular change observed in stress-reactive group was the increase in UCN2 plasma level. The differentiated behavioral stress responses were reflected by gene expression changes in the pituitary. Alterations in the mRNA levels of Crh, Ucn2 and Ucn3 in the pituitary might confirm the paracrine and/or autocrine effects of these peptides in stress response. The opposite behavioral effect between SNR vs. SIR groups and the surprising similarity at gene expression and plasma ACTH levels in these two groups may suggest the discrepancy between molecular and behavioral stress responses; however, there results might indicate to similarity underlying different ways to cope with stress conditions.


Assuntos
Comunicação Autócrina , Regulação da Expressão Gênica , Neuropeptídeos/sangue , Comunicação Parácrina , Hipófise/metabolismo , Estresse Fisiológico , Animais , Comportamento Animal , Masculino , Hipófise/patologia , Ratos , Ratos Wistar
19.
Psychopharmacology (Berl) ; 227(4): 583-93, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23377023

RESUMO

RATIONALE: Few studies have investigated neurobiological and biochemical differences between stress-resilient and stress-vulnerable experimental animals. OBJECTIVES: We investigated alterations in mesolimbic dopamine D2 receptor density and mRNA expression level in stressed rats at two time points, i.e. after 2 and 5 weeks of chronic mild stress (CMS). METHODS: We used the chronic mild stress paradigm because it is a well-established animal model of depression. Two groups of stressed rats were distinguished during CMS experiments: (1) stress reactive (70 %), which displayed a decrease in the drinking of a palatable sucrose solution during the stress regimen, and (2) stress resilient (30 %), which exhibited an unaltered drinking profile when compared with the unchallenged control group. [(3)H]Domperidone was used as a ligand to label dopamine D2 receptors, and a mixture of three specific oligonucleotides was used to evaluate dopamine D2 receptor mRNA changes in various regions of the rat brain. RESULTS: CMS strongly affected the mesolimbic dopamine circuit in stress-resilient group after 2 weeks and stress-reactive group of rats after 5 weeks which exhibited a decrease in the level of dopamine D2 receptor protein without alterations in D2 mRNA expression. Stress-resilient animals, but not stress-reactive animals, effectively adapted to the extended stress and coped with it. The increase in D2 mRNA expression returned the dopamine D2 receptor density to control levels in stress-resilient rats after 5 weeks of CMS, but not in stress-reactive animals. CONCLUSIONS: These results clearly demonstrate that, despite earlier blunting, the activation of dopamine receptor biosynthesis in the dopamine mesoaccumbens system in stress-resilient rats is involved in active coping with stressful experiences, and it exhibits a delay in time.


Assuntos
Encéfalo/metabolismo , Depressão/fisiopatologia , Receptores de Dopamina D2/metabolismo , Estresse Psicológico/fisiopatologia , Animais , Modelos Animais de Doenças , Domperidona/metabolismo , Regulação da Expressão Gênica , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Dopamina D2/genética , Sacarose/administração & dosagem , Fatores de Tempo
20.
Pharmacol Rep ; 65(6): 1498-505, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24552997

RESUMO

G protein-coupled receptors (GPCRs) represent the largest family of membrane proteins in the human genome and are the target of approximately half of all therapeutic drugs. For many years, GPCRs were thought to exist and function as monomeric units. However, during the past two decades, substantial biochemical, structural and functional evidence have indicated that GPCRs can associate and form heteromers that exhibit functional properties distinct from the corresponding monomers. The understanding of the unique pharmacological and functional properties of such heteromers is a major challenge for neuroscience, particularly given the abundant evidence suggesting that GPCR heteromers may play a crucial role in neuropsychiatric disorders. Herein, we present current data on the role of GPCR heterodimerization in neuropsychiatric disorders, with a focus on its potential implications in depression. The presented examples of pairs of receptors, with their specific pharmacological and functional properties, are likely to lead to novel effective strategies in antidepressant drug development. The currently available techniques for studying GPCR heterodimerization, both in vitro as well as in situ in native tissue, are also described.


Assuntos
Depressão/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Dimerização , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA