Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Neurochem Res ; 49(8): 2179-2196, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38834845

RESUMO

There is some evidence that the serotonin receptor subtype 7 (5-HT7) could be new therapeutic target for neuroprotection. The aim of this study was to compare the neuroprotective and neurite outgrowth potential of new 5-HT7 receptor agonists (AH-494, AGH-238, AGH-194) with 5-CT (5-carboxyamidotryptamine) in human neuroblastoma SH-SY5Y cells. The results revealed that 5-HT7 mRNA expression was significantly higher in retinoic acid (RA)-differentiated cells when compared to undifferentiated ones and it was higher in cell cultured in neuroblastoma experimental medium (DMEM) compared to those placed in neuronal (NB) medium. Furthermore, the safety profile of compounds was favorable for all tested compounds at concentration used for neuroprotection evaluation (up to 1 µM), whereas at higher concentrations (above 10 µM) the one of the tested compounds, AGH-194 appeared to be cytotoxic. While we observed relatively modest protective effects of 5-CT and AH-494 in UN-SH-SY5Y cells cultured in DMEM, in UN-SH-SY5Y cells cultured in NB medium we found a significant reduction of H2O2-evoked cell damage by all tested 5-HT7 agonists. However, 5-HT7-mediated neuroprotection was not associated with inhibition of caspase-3 activity and was not observed in RA-SH-SY5Y cells exposed to H2O2. Furthermore, none of the tested 5-HT7 agonists altered the damage induced by 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenylpyridinium ion (MPP +) and doxorubicin (Dox) in UN- and RA-SH-SY5Y cells cultured in NB. Finally we showed a stimulating effect of AH-494 and AGH-194 on neurite outgrowth. The obtained results provide insight into neuroprotective and neurite outgrowth potential of new 5-HT7 agonists.


Assuntos
Neuroblastoma , Crescimento Neuronal , Fármacos Neuroprotetores , Receptores de Serotonina , Agonistas do Receptor de Serotonina , Humanos , Receptores de Serotonina/metabolismo , Fármacos Neuroprotetores/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Linhagem Celular Tumoral , Neuroblastoma/patologia , Neuroblastoma/metabolismo , Crescimento Neuronal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sobrevivência Celular/efeitos dos fármacos , Serotonina/análogos & derivados
2.
J Neurochem ; 166(3): 623-632, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37358014

RESUMO

Prediction of post-stroke depressive symptoms (DSs) is challenging in patients without a history of depression. Gene expression profiling in blood cells may facilitate the search for biomarkers. The use of an ex vivo stimulus to the blood helps to reveal differences in gene profiles by reducing variation in gene expression. We conducted a proof-of-concept study to determine the usefulness of gene expression profiling in lipopolysaccharide (LPS)-stimulated blood for predicting post-stroke DS. Out of 262 enrolled patients with ischemic stroke, we included 96 patients without a pre-stroke history of depression and not taking any anti-depressive medication before or during the first 3 months after stroke. We assessed DS at 3 months after stroke using the Patient Health Questionnaire-9. We used RNA sequencing to determine the gene expression profile in LPS-stimulated blood samples taken on day 3 after stroke. We constructed a risk prediction model using a principal component analysis combined with logistic regression. We diagnosed post-stroke DS in 17.7% of patients. Expression of 510 genes differed between patients with and without DS. A model containing 6 genes (PKM, PRRC2C, NUP188, CHMP3, H2AC8, NOP10) displayed very good discriminatory properties (area under the curve: 0.95) with the sensitivity of 0.94 and specificity of 0.85. Our results suggest the potential utility of gene expression profiling in whole blood stimulated with LPS for predicting post-stroke DS. This method could be useful for searching biomarkers of post-stroke depression.


Assuntos
Lipopolissacarídeos , Acidente Vascular Cerebral , Humanos , Lipopolissacarídeos/farmacologia , Depressão/genética , Acidente Vascular Cerebral/complicações , Perfilação da Expressão Gênica , Biomarcadores , Complexos Endossomais de Distribuição Requeridos para Transporte
3.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36293185

RESUMO

The expression of the Calcium/Calmodulin-Dependent Protein Kinase I gamma (encoded by the Camk1g gene) depends on the activation of glucocorticoid receptors (GR) and is strongly regulated by stress. Since Camk1g is primarily expressed in neuronal cells of the limbic system in the brain, we hypothesized that it could be involved in signaling mechanisms that underlie the adaptive or maladaptive responses to stress. Here, we find that restraint-induced stress and the GR agonist dexamethasone robustly increase the expression of Camk1g in neurons of the amygdalar nuclei in the mouse brain. To assess the functional role of Camk1g expression, we performed a virally induced knock-down of the transcript. Mice with bilateral amygdala-specific Camk1g knock-down showed increased anxiety-like behaviors in the light-dark box, and an increase in freezing behavior after fear-conditioning, but normal spatial working memory during exploration of a Y-maze. Thus, we confirm that Camk1g is a neuron-specific GR-regulated transcript, and show that it is specifically involved in behaviors related to anxiety, as well as responses conditioned by aversive stimuli.


Assuntos
Núcleo Central da Amígdala , Glucocorticoides , Camundongos , Animais , Glucocorticoides/farmacologia , Núcleo Central da Amígdala/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Cálcio , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/metabolismo , Ansiedade/metabolismo , Dexametasona/farmacologia , Comportamento Animal
4.
J Transl Med ; 17(1): 141, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31046777

RESUMO

BACKGROUND: Rupture of an intracranial aneurysm (IA) causes a systemic response that involves an immune/inflammatory reaction. Our previous study revealed a downregulation of genes related to T lymphocytes and an upregulation of genes related to monocytes and neutrophils after IA rupture. It remains unknown whether that resulted from alterations in transcription or cell count. We sought to characterize the systemic response to IA rupture through analysis of transcript expression profiles in peripheral blood cells. We also investigated effects of IA rupture on the composition of mononuclear cells in peripheral blood. METHODS: We included 19 patients in the acute phase of IA rupture (RAA, first 72 h), 20 patients in the chronic phase (RAC, 3-15 months), and 20 controls. Using deep transcriptome sequencing, we analyzed the expression of protein-coding and noncoding RNAs. Expression levels, transcript biotypes, alternative splicing and other features of the regulated transcripts were studied. A functional analysis was performed to determine overrepresented ontological groups among gene expression profiles. Flow cytometry was used to analyze alterations in the level of mononuclear leukocyte subpopulations. RESULTS: Comparing RAA and controls, we identified 491 differentially expressed transcripts (303 were downregulated, and 188 were upregulated in RAA). The results indicate that the molecular changes in response to IA rupture occur at the level of individual transcripts. Functional analysis revealed that the most impacted biological processes are related to regulation of lymphocyte activation and toll-like receptor signaling pathway. Differences between RAC and controls were less prominent. Analysis of leukocyte subsets revealed a significantly decreased number of CD4+ lymphocytes and increase of classical and intermediate monocytes in RAA patients compared to controls. CONCLUSIONS: IA rupture in the acute phase strongly influences the transcription profiles of peripheral blood cells as well as the composition of mononuclear cells. A specific pattern of gene expression alteration was found, suggesting a depression of lymphocyte response and enhancement of monocyte activity.


Assuntos
Aneurisma Roto/genética , Regulação da Expressão Gênica , Aneurisma Intracraniano/genética , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Anotação de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Transcrição Gênica , Transcriptoma/genética
5.
BMC Neurosci ; 18(1): 37, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28381250

RESUMO

BACKGROUND: The mechanisms of steroids actions in the brain mainly involve the binding and nuclear translocation of specific cytoplasmic receptors. These receptors can act as transcription factors and regulate gene expression. However, steroid-dependent transcriptional regulation in different types of neural cells is not yet fully understood. The aim of this study was to evaluate and compare transcriptional alterations induced by various steroid receptor agonists in primary cultures of astrocytes and neurons from mouse brain. RESULTS: We utilized whole-genome microarrays (Illumina Mouse WG-6) and quantitative PCR analyses to measure mRNA abundance levels. To stimulate gene expression we treated neuronal and astroglial cultures with dexamethasone (100 nM), aldosterone (200 nM), progesterone (200 nM), 5α-dihydrotestosterone (200 nM) and ß-Estradiol (200 nM) for 4 h. Neurons were found to exhibit higher levels of expression of mineralocorticoid receptor, progesterone receptor and estrogen receptor 2 than astrocytes. However, higher mRNA level of glucocorticoid receptor mRNA was observed in astrocytes. We identified 956 genes regulated by steroids. In astrocytes we found 381 genes altered by dexamethasone and 19 altered by aldosterone. Functional classification of the regulated genes indicated their putative involvement in multiple aspects of cell metabolism (up-regulated Slc2a1, Pdk4 and Slc45a3) and the inflammatory response (down-regulated Ccl3, Il1b and Tnf). Progesterone, dihydrotestosterone and estradiol did not change gene expression in astrocytes. We found no significant changes in gene expression in neurons. CONCLUSIONS: The obtained results indicate that glial cells might be the primary targets of transcriptional action of steroids in the central nervous system. Substantial changes in gene expression driven by the glucocorticoid receptor imply an important role for the hypothalamic-pituitary-adrenal axis in the hormone-dependent regulation of brain physiology. This is an in vitro study. Hence, the model may not accurately reflect all the effects of steroids on gene expression in neurons in vivo.


Assuntos
Astrócitos/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de Esteroides/agonistas , Esteroides/farmacologia , Transcriptoma/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Células Cultivadas , Análise por Conglomerados , Corpo Estriado/metabolismo , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Camundongos Endogâmicos C57BL , Análise em Microsséries , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Receptores de Esteroides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma/fisiologia
6.
Gene ; : 148890, 2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39187136

RESUMO

Oprm1, the gene encoding the µ-opioid receptor, has multiple reported transcripts, with a variable 3' region and many alternative sequences encoding the C-terminus of the protein. The functional implications of this variability remain mostly unexplored, though a recurring notion is that it could be exploited by developing selective ligands with improved clinical profiles. Here, we comprehensively examined Oprm1 transcriptional variants in the murine central nervous system, using long-read RNAseq as well as spatial and single-cell transcriptomics. The results were validated with RNAscope in situ hybridization. We found a mismatch between transcripts annotated in the mouse genome (GRCm38/mm10) and the RNA-seq results. Sequencing data indicated that the primary Oprm1 transcript has a 3' terminus located on chr10:6,860,027, which is ∼ 9.5 kilobases downstream of the longest annotated exon 4 end. Long-read sequencing confirmed that the final Oprm1 exon included a 10.2 kilobase long 3' untranslated region, and the presence of the long variant was unambiguously confirmed using RNAscope in situ hybridization in the thalamus, striatum, cortex and spinal cord. Conversely, expression of the Oprm1 reference transcript or alternative transcripts of the Oprm1 gene was absent or close to the detection limit. Thus, the primary transcript of the Oprm1 mouse gene is a variant with a long 3' untranslated region, which is homologous to the human OPRM1 primary transcript and encodes the same conserved C-terminal amino acid sequence.

7.
Glia ; 61(4): 623-35, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23339081

RESUMO

Chronic opioid use leads to the structural reorganization of neuronal networks, involving genetic reprogramming in neurons and glial cells. Our previous in vivo studies have revealed that a significant fraction of the morphine-induced alterations to the striatal transcriptome included glucocorticoid (GC) receptor (GR)-dependent genes. Additional analyses suggested glial cells to be the locus of these changes. In the current study, we aimed to differentiate the direct transcriptional effects of morphine and a GR agonist on primary striatal neurons and astrocytes. Whole-genome transcriptional profiling revealed that while morphine had no significant effect on gene expression in both cell types, dexamethasone significantly altered the transcriptional profile in astrocytes but not neurons. We obtained a complete dataset of genes undergoing the regulation, which includes genes related to glucose metabolism (Pdk4), circadian activity (Per1) and cell differentiation (Sox2). There was also an overlap between morphine-induced transcripts in striatum and GR-dependent transcripts in cultured astrocytes. We further analyzed the regulation of expression of one gene belonging to both groups, serum and GC regulated kinase 1 (Sgk1). We identified two transcriptional variants of Sgk1 that displayed selective GR-dependent upregulation in cultured astrocytes but not neurons. Moreover, these variants were the only two that were found to be upregulated in vivo by morphine in a GR-dependent fashion. Our data suggest that the morphine-induced, GR-dependent component of transcriptome alterations in the striatum is confined to astrocytes. Identification of this mechanism opens new directions for research on the role of astrocytes in the central effects of opioids.


Assuntos
Astrócitos/metabolismo , Marcação de Genes/métodos , Morfina/administração & dosagem , Neurônios/fisiologia , Receptores de Glucocorticoides/fisiologia , Transdução de Sinais/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
Int J Neuropsychopharmacol ; 15(10): 1457-71, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22040728

RESUMO

The brain's noradrenergic system is involved in the development of behaviours induced by drugs of abuse, e.g. dependence and withdrawal, and also reward or psychomotor effects. To investigate how noradrenergic system activity is controlled in the context associated with drug-induced behaviours, we generated a Cre/loxP mouse model in which the essential glutamate NMDA receptor subunit NR1 is ablated in cells expressing dopamine ß-hydroxylase (Dbh). As a result, the noradrenergic cells in NR1DbhCre mice lack the NMDA receptor-dependent component of excitatory post-synaptic currents. The mutant mice displayed no obvious behavioural alterations, had unchanged noradrenaline content and mild increase in dopamine levels in the nucleus accumbens. Interestingly, NR1DbhCre animals did not develop morphine-induced psychomotor sensitization. However, when the morphine injections were preceded by treatment with RX821002, an antagonist of α2-adrenergic receptors, the development of sensitization was restored. Conversely, pretreatment with clonidine, an agonist of α2-adrenergic receptors, blocked development of sensitization in wild-type mice. We also found that while the development of tolerance to morphine was normal in mutant mice, withdrawal symptoms were attenuated. These data reveal that NMDA receptors on noradrenergic neurons regulate development of opiate dependence and psychomotor sensitization, by controlling drug-induced noradrenaline signalling.


Assuntos
Neurônios Adrenérgicos/metabolismo , Proteínas de Transporte/biossíntese , Ácido Glutâmico/fisiologia , Dependência de Morfina/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Desempenho Psicomotor/fisiologia , Neurônios Adrenérgicos/efeitos dos fármacos , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dependência de Morfina/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Técnicas de Cultura de Órgãos , Desempenho Psicomotor/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato
9.
Cells ; 11(16)2022 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-36010662

RESUMO

Despite the general awareness of the need to reduce air pollution, the efforts were undertaken in Poland to eliminate the pollutants and their harmful effect on human health seem to be insufficient. Moreover, the latest data indicate that the city of Krakow is at the forefront of the most polluted cities worldwide. Hence, in this report, we investigated the impact of particulate matter isolated from the air of Krakow (PM KRK) on the gene expression profile of peripheral blood mononuclear cells (PBMCs) in healthy donors (HD) and patients with atherosclerosis (AS), rheumatoid arthritis (RA) and multiple sclerosis (MS), after in vitro exposure. Blood samples were collected in two seasons, differing in the concentration of PM in the air (below or above a daily limit of 50 µg/m3 for PM 10). Data show that PBMCs exposed in vitro to PM KRK upregulated the expression of genes involved, among others, in pro-inflammatory response, cell motility, and regulation of cell metabolism. The transcriptional effects were observed predominantly in the group of patients with AS and MS. The observed changes seem to be dependent on the seasonal concentration of PM in the air of Krakow and may suggest their important role in the progression of AS, MS, and RA in the residents of Krakow.


Assuntos
Poluentes Atmosféricos , Doenças Autoimunes , Humanos , Leucócitos Mononucleares , Tamanho da Partícula , Smog
10.
Transl Psychiatry ; 11(1): 246, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33903586

RESUMO

Altered cytokine synthesis thought to contribute to the pathophysiology of post-stroke depression (PSD). Toll-like receptor 4 (TLR4) is a master regulator of innate immunity. The aim of this study was to explore the putative association between TLR4-mediated cytokine synthesis and subsequent symptoms of PSD. In total, 262 patients with ischemic stroke and without a history of PSD were included. Depressive symptoms were assessed using the Patient Health Questionnaire-9 in 170 patients on Day 8 and in 146 at 3 months after stroke. Blood samples taken on Day 3 after stroke were stimulated ex vivo with lipopolysaccharide (LPS). Ex vivo synthesized cytokines (TNFα, IP-10, IL-1ß, IL-6, IL-8, IL-10, and IL-12p70) and circulating cytokines (TNFα, IL-6, sIL-6R, and IL-1ra) were measured using the enzyme-linked immunoassay or cytometric method. RNA sequencing was used to determine the gene expression profile of LPS-induced cytokines and chemokines. LPS-induced cytokine synthesis and the gene expression of TLR4-dependent cytokines and chemokines did not differ between patients with and without greater depressive symptoms. The plasma level of IL-6, but not TNFα, sIL-6R, and IL-1ra, was higher in patients who developed depressive symptoms at 3 months after stroke (median: 4.7 vs 3.4 pg/mL, P = 0.06). Plasma IL-6 predicted the severity of depressive symptoms at 3 months after stroke (ß = 0.42, P = 0.03). In conclusion, TLR4-dependent cytokine synthesis was not associated with greater post-stroke depressive symptoms in this study. Circulating IL-6 might be associated with depressive symptoms occurring at 3 months after stroke.


Assuntos
Acidente Vascular Cerebral , Receptor 4 Toll-Like , Citocinas , Depressão/etiologia , Expressão Gênica , Humanos , Lipopolissacarídeos , Acidente Vascular Cerebral/complicações
11.
J Mol Med (Berl) ; 98(6): 887-896, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32424559

RESUMO

Multiple classes of small RNAs (sRNAs) are expressed in the blood and are involved in the regulation of pivotal cellular processes. We aimed to elucidate the expression patterns and functional roles of sRNAs in the systemic response to intracranial aneurysm (IA) rupture. We used next-generation sequencing to analyze the expression of sRNAs in patients in the acute phase of IA rupture (first 72 h), in the chronic phase (3-15 months), and controls. The patterns of alterations in sRNA expression were analyzed in the context of clinically relevant information regarding the biological consequences of IA rupture. We identified 542 differentially expressed sRNAs (108 piRNAs, 99 rRNAs, 90 miRNAs, 43 scRNAs, 36 tRNAs, and 32 snoRNAs) among the studied groups with notable differences in upregulated and downregulated sRNAs between the groups and sRNAs categories. piRNAs and rRNAs showed a substantial decrease in RNA abundance that was sustained after IA rupture, whereas miRNAs were largely upregulated. Downregulated sRNA genes included piR-31080, piR-57947, 5S rRNA, LSU-rRNA, and SSU-rRNA s. Remarkable enrichment in the representation of transcription factor binding sites was revealed in genomic locations of the regulated sRNA. We found strong overrepresentation of glucocorticoid receptor, retinoid x receptor alpha, and estrogen receptor alpha binding sites at the locations of downregulated piRNAs, tRNAs, and rRNAs. This report, although preliminary and largely proof-of-concept, is the first to describe alterations in sRNAs abundance levels in response to IA rupture in humans. The obtained results indicate novel mechanisms that may constitute another level of control of the inflammatory response. KEY MESSAGES: A total of 542 sRNAs were differentially expressed after aneurysmal SAH comparing with controls piRNAs and rRNAs were upregulated and miRNAs were downregulated after IA rupture The regulated sRNA showed an enrichment in the representation of some transcription factor binding sites piRNAs, tRNAs, and rRNAs showed an overrepresentation for GR, RXRA, and ERALPHA binding sites.


Assuntos
Biomarcadores , Ácidos Nucleicos Livres , MicroRNAs/sangue , RNA Ribossômico/sangue , RNA Interferente Pequeno/sangue , Hemorragia Subaracnóidea/sangue , Adulto , Suscetibilidade a Doenças , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Hemorragia Subaracnóidea/diagnóstico , Hemorragia Subaracnóidea/etiologia , Fatores de Transcrição/metabolismo
12.
Mol Neurobiol ; 57(2): 988-996, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31654316

RESUMO

Influence of an intracranial aneurysm (IA) rupture on the expression of miRNAs and the potential significance of the resulting changes remains poorly understood. We aimed to characterize the response to the IA rupture through the analysis of miRNAs in peripheral blood cells. Expression of small RNAs was investigated using deep transcriptome sequencing in patients in the acute phase of an IA rupture (first 72 h), in the chronic phase (3-15 months), and controls. A functional analysis and the potential interactions between miRNAs and target genes were investigated. We also measured the levels of proteins that were influenced by regulated miRNAs. We found that 106 mature miRNAs and 90 miRNA precursors were differentially expressed among the groups. The regulated miRNAs were involved in a variety of pathways, and the top pathway involved cytokine-cytokine receptor interactions. The identified miRNAs targeted the inflammatory factors HMGB1 and FASLG. Changes in their expression were detected at the mRNA and protein levels. IA rupture strongly influences the transcription profiles in peripheral blood cells. The regulated miRNAs were involved in the control of immune cell homeostasis. In summary, these results may aid in the elucidation of the molecular mechanisms that orchestrate the inflammatory response to IA rupture.


Assuntos
Inflamação/metabolismo , Aneurisma Intracraniano/metabolismo , Aneurisma Intracraniano/patologia , MicroRNAs/metabolismo , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Inflamação/genética , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Mensageiro/genética , Transcriptoma/fisiologia
13.
Neuropharmacology ; 180: 108307, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32941853

RESUMO

Opioid signaling controls the activity of the brain's reward system. It is involved in signaling the hedonic effects of rewards and has essential roles in reinforcement and motivational processes. Here, we focused on opioid signaling through mu and delta receptors on dopaminoceptive neurons and evaluated the role these receptors play in reward-driven behaviors. We generated a genetically modified mouse with selective double knockdown of mu and delta opioid receptors in neurons expressing dopamine receptor D1. Selective expression of the transgene was confirmed using immunostaining. Knockdown was validated by measuring the effects of selective opioid receptor agonists on neuronal membrane currents using whole-cell patch clamp recordings. We found that in the nucleus accumbens of control mice, the majority of dopamine receptor D1-expressing neurons were sensitive to a mu or delta opioid agonist. In mutant mice, the response to the delta receptor agonist was blocked, while the effects of the mu agonist were strongly attenuated. Behaviorally, the mice had no obvious impairments. The mutation did not affect the sensitivity to the rewarding effects of morphine injections or social contact and had no effect on preference for sweet taste. Knockdown had a moderate effect on motor activity in some of the tests performed, but this effect did not reach statistical significance. Thus, we found that knocking down mu and delta receptors on dopamine receptor D1-expressing cells does not appreciably affect some of the reward-driven behaviors previously attributed to opioid signaling.


Assuntos
Neurônios/metabolismo , Receptores de Dopamina D1/biossíntese , Receptores Opioides delta/deficiência , Receptores Opioides mu/deficiência , Recompensa , Analgésicos Opioides/farmacologia , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Receptores de Dopamina D1/genética , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
14.
Neuropsychopharmacology ; 45(2): 404-415, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31254970

RESUMO

To date, neurons have been the primary focus of research on the role of glucocorticoids in the regulation of brain function and pathological behaviors, such as addiction. Astrocytes, which are also glucocorticoid-responsive, have been recently implicated in the development of drug abuse, albeit through as yet undefined mechanisms. Here, using a spectrum of tools (whole-transcriptome profiling, viral-mediated RNA interference in vitro and in vivo, behavioral pharmacology and electrophysiology), we demonstrate that astrocytes in the nucleus accumbens (NAc) are an important locus of glucocorticoid receptor (GR)-dependent transcriptional changes that regulate rewarding effects of morphine. Specifically, we show that targeted knockdown of the GR in the NAc astrocytes enhanced conditioned responses to morphine, with a concomitant inhibition of morphine-induced neuronal excitability and plasticity. Interestingly, GR knockdown did not influence sensitivity to cocaine. Further analyses revealed GR-dependent regulation of astroglial metabolism. Notably, GR knockdown inhibited induced by glucocorticoids lactate release in astrocytes. Finally, lactate administration outbalanced conditioned responses to morphine in astroglial GR knockdown mice. These findings demonstrate a role of GR-dependent regulation of astrocytic metabolism in the NAc and a key role of GR-expressing astrocytes in opioid reward processing.


Assuntos
Analgésicos Opioides/farmacologia , Astrócitos/metabolismo , Condicionamento Psicológico/fisiologia , Ácido Láctico/metabolismo , Morfina/farmacologia , Receptores de Glucocorticoides/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Condicionamento Psicológico/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
15.
Am J Pathol ; 173(4): 981-92, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18787101

RESUMO

We investigated the role of heme oxygenase-1 (HO-1), a powerful anti-inflammatory and anti-oxidant enzyme, in modulating cigarette smoke (CS)-induced mucus secretion. In both rats and mice, 5-day CS exposure increased HO-1 expression and activity, mucus secretion, MUCIN 5AC (MUC5AC) gene and protein expression, and local inflammation, along with up-regulation of dual oxidase 1 gene expression and both the activity and phosphorylation of the epidermal growth factor receptor, which is involved in MUC5AC induction. Pharmacological induction of HO-1 prevented these actions and inhibition of HO-1 expression by a specific siRNA potentiated them. In French participants to the European Community Respiratory Health Survey II (n = 210, 30 to 53 years of age, 50% males) exposed to CS, a significant increase in the percentage of participants with chronic sputum was observed in those harboring at least one allele with a long (GT)(n) in the HO-1 promoter gene (>33 repeats), which is associated with a low level of HO-1 protein expression, compared with those with a short number of (GT)n repeats (21.7% versus 8.6%, P = 0.047). No such results were observed in those who had never smoked (n = 297). We conclude that HO-1 has a significant protective effect against airway mucus hypersecretion in animals and humans exposed to CS.


Assuntos
Heme Oxigenase-1/metabolismo , Pulmão/enzimologia , Pulmão/metabolismo , Muco/metabolismo , Fumar/efeitos adversos , Adulto , Animais , Regulação para Baixo/efeitos dos fármacos , Oxidases Duais , Indução Enzimática/efeitos dos fármacos , Feminino , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Hemina/farmacologia , Humanos , Técnicas In Vitro , Inflamação , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mucina-5AC , Mucinas/metabolismo , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Regiões Promotoras Genéticas/genética , Ratos , Ratos Sprague-Dawley , Sequências Repetitivas de Ácido Nucleico/genética , Escarro/enzimologia
16.
J Clin Med ; 8(11)2019 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-31703409

RESUMO

: We sought to investigate whether systematic balance training modulates brain area activity responsible for postural control and influence brain-derived neurotrophic factor (BDNF) mRNA protein expression. Seventy-four older adults were randomly divided into three groups (mean age 65.34 ± 3.79 years, 30 females): Classic balance exercises (CBT), virtual reality balance exercises (VBT), and control (CON). Neuroimaging studies were performed at inclusion and after completion of the training or 12 weeks later (CON). Blood samples were obtained to measure BDNF expression. The study revealed significant interaction of sessions and groups: In the motor imagery (MI) condition for supplementary motor area (SMA) activity (Fat peak = 5.25, p < 0.05); in the action observation (AO) condition for left and right supramarginal gyrus/posterior insula (left: Fat peak = 6.48, p < 0.05; right: Fat peak = 6.92, p < 0.05); in the action observation together with motor imagery (AOMI) condition for the middle occipital gyrus (laterally)/area V5 (left: Fat peak = 6.26, p < 0.05; right: Fat peak = 8.37, p < 0.05), and in the cerebellum-inferior semilunar lobule/tonsil (Fat peak = 5.47, p < 0.05). After the training serum BDNF level has increased in CBT (p < 0.001) and in CBT compared to CON (p < 0.05). Systematic balance training may reverse the age-related cortical over-activations and appear to be a factor mediating neuroplasticity in older adults.

17.
Cancer Lett ; 432: 1-16, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-29885518

RESUMO

The present study aimed to determine the role of metabotropic glutamate receptor 8 (mGluR8) in tumor biology. Using various molecular approaches (RNAi or GRM8 cDNA), cell clones with downregulated (human neuroblastoma SH-SY5Y and human glioma LN229) or overexpressed (human glioma U87-MG and LN18 cell lines) mGluR8 were generated. Next, comparative studies on cell proliferation and migration rates, induction of apoptosis and chemosensitivity were performed among these clones. The mGluR8-downregulated SH-SY5Y clones proliferated faster and were more resistant to cytotoxic action of staurosporine, doxorubicin, irinotecan and cisplatin when compared to control cells. Moreover, these clones were characterized by a lower activity of caspases, calpains and some kinases (GSK-3ß, Akt and JNK). The mGluR8-downregulated LN229 clones migrated faster and were less prone to cell-damaging effect of staurosporine and irinotecan when compared with relevant control cells. In contrast, in GRM8-overexpressing U87-MG and LN18 clones, a decreased cell proliferation, increased apoptosis and elevated vulnerability to some cytotoxic agents were found. Altogether, our in vitro data for the first time evidenced a tumor suppressor and chemosensitizing role of mGluR8.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Glioma/patologia , Neuroblastoma/patologia , Receptores de Glutamato Metabotrópico/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Movimento Celular , Expressão Ectópica do Gene , Regulação Neoplásica da Expressão Gênica , Glioma/tratamento farmacológico , Glioma/metabolismo , Humanos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Receptores de Glutamato Metabotrópico/genética , Células Tumorais Cultivadas
18.
Neuropharmacology ; 141: 223-237, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30170084

RESUMO

A specific activation of metabotropic glutamate receptor 7 (mGluR7) has been shown to be neuroprotective in various models of neuronal cell damage, however, its role in glia cell survival has not been studied, yet. Thus, we performed comparative experiments estimating protective effects of the mGluR7 allosteric agonist AMN082 in glia, neuronal and neuronal-glia cell cultures against various harmful stimuli. First, the transcript levels of mGluR7 and other subtypes of group II and III mGluRs in cortical neuronal, neuronal-glia and glia cell cultures have been measured by qPCR method. Next, we demonstrated that AMN082 with similar efficiency attenuated the glia cell damage evoked by staurosporine (St) and doxorubicin (Dox). The AMN082-mediated glioprotection was mGluR7-dependent and associated with decreased DNA fragmentation without involvement of caspase-3 inhibition. Moreover, the inhibitors of PI3K/Akt and MAPK/ERK1/2 pathways blocked the protective effect of AMN082. In neuronal and neuronal-glia cell cultures in the model of glutamate (Glu)- but not St-evoked cell damage, we showed a significant glia contribution to mGluR7-mediated neuroprotection. Finally, by using glia and neuronal cells derived from mGluR7+/+ and mGluR7-/- mice we demonstrated a higher cell-damaging effect of St and Dox in mGluR7-deficient glia but not in neurons (cerebellar granule cells). Our present data showed for the first time a glioprotective potential of AMN082 underlain by mechanisms involving the activation of PI3K/Akt and MAPK/ERK1/2 pathways and pro-survival role of mGluR7 in glia cells. These findings together with the confirmed neuroprotective properties of AMN082 justify further research on mGluR7-targeted therapies for various CNS disorders.


Assuntos
Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/agonistas , Animais , Compostos Benzidrílicos/antagonistas & inibidores , Caspase 3/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/metabolismo , Técnicas de Cocultura , Fragmentação do DNA/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Doxorrubicina/antagonistas & inibidores , Inibidores Enzimáticos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de Glutamato Metabotrópico/genética , Transdução de Sinais , Estaurosporina/antagonistas & inibidores
19.
Transl Psychiatry ; 8(1): 255, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30487639

RESUMO

Stress elicits the release of glucocorticoids (GCs) that regulate energy metabolism and play a role in emotional memory. Astrocytes express glucocorticoid receptors (GR), but their contribution to cognitive effects of GC's action in the brain is unknown. To address this question, we studied how astrocyte-specific elimination of GR affects animal behavior known to be regulated by stress. Mice with astrocyte-specific ablation of GR presented impaired aversive memory expression in two different paradigms of Pavlovian learning: contextual fear conditioning and conditioned place aversion. These mice also displayed compromised regulation of genes encoding key elements of the glucose metabolism pathway upon GR stimulation. In particular, we identified that the glial, but not the neuronal isoform of a crucial stress-response molecule, Sgk1, undergoes GR-dependent regulation in vivo and demonstrated the involvement of SGK1 in regulation of glucose uptake in astrocytes. Together, our results reveal astrocytes as a central element in GC-dependent formation of aversive memory and suggest their relevance for stress-induced alteration of brain glucose metabolism. Consequently, astrocytes should be considered as a cellular target of therapies of stress-induced brain diseases.


Assuntos
Astrócitos/metabolismo , Comportamento Animal/fisiologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Memória/fisiologia , Nociceptividade/fisiologia , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/fisiologia , Estresse Psicológico/metabolismo , Animais , Proteínas Imediatamente Precoces/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/metabolismo
20.
Free Radic Biol Med ; 40(7): 1250-63, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16545694

RESUMO

BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Recently the involvement of HO-1 in angiogenesis has been shown; however, the role of heme and HO-1 in wound healing angiogenesis has not been yet investigated. RESULTS: Treatment of HaCaT keratinocytes with hemin (heme chloride) induced HO-1 expression and activity. The effect of heme on vascular endothelial growth factor (VEGF) synthesis is variable: induction is significant after a short, 6 h treatment with heme, while longer stimulation may attenuate its production. The involvement of HO-1 in VEGF synthesis was confirmed by inhibition of VEGF expression by SnPPIX, a blocker of HO activity and by attenuation of HO-1 mRNA expression with specific siRNA. Importantly, induction of HO-1 by hemin was able to overcome the inhibitory effect of high glucose on VEGF synthesis. Moreover, HO-1 expression was also induced in keratinocytes cultured in hypoxia, with concomitant augmentation of VEGF production, which was further potentiated by hemin stimulation. Accordingly, conditioned media from keratinocytes overexpressing HO-1 enhanced endothelial cell proliferation and augmented formation of capillaries in angiogenic assay in vitro. CONCLUSIONS: HO-1 is involved in hemin-induced VEGF expression in HaCaT and may play a role in hypoxic regulation of this protein. HO-1 overexpression may be beneficial in restoring the proper synthesis of VEGF disturbed in diabetic conditions.


Assuntos
Heme Oxigenase-1/farmacologia , Hemina/farmacologia , Queratinócitos/metabolismo , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Endotélio Vascular/citologia , Radicais Livres/metabolismo , Glucose/farmacologia , Humanos , Hipóxia/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA