Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 164(5): 598-612, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36161462

RESUMEN

Noradrenergic neurotransmission is a critical mediator of stress responses. In turn, exposure to stress induces noradrenergic system adaptations, some of which are implicated in the etiology of stress-related disorders. Adrenergic receptors (ARs) in the ventral tegmental area (VTA) have been demonstrated to regulate phasic dopamine (DA) release in the forebrain, necessary for behavioral responses to conditional cues. However, the impact of stress on noradrenergic modulation of the VTA has not been previously explored. We demonstrate that ARs in the VTA regulate dopaminergic activity in the VTA-BLA (basolateral amygdala) circuit, a key system for processing stress-related stimuli; and that such control is altered by acute stress. We utilized fast-scan cyclic voltammetry to assess the effects of intra-VTA microinfusion of α1 -AR and α2 -AR antagonists (terazosin and RX-821002, respectively), on electrically evoked phasic DA release in the BLA in stress-naïve and stressed (unavoidable electric shocks - UES) anesthetized male Sprague-Dawley rats. In addition, we used western blotting to explore UES-induced alterations in AR protein level in the VTA. Intra-VTA terazosin or RX-821002 dose-dependently attenuated DA release in the BLA. Interestingly, UES decreased the effects of intra-VTA α2 -AR blockade on DA release (24 h but not 7 days after stress), while the effects of terazosin were unchanged. Despite changes in α2 -AR physiological function in the VTA, UES did not alter α2 -AR protein levels in either intracellular or membrane fractions. These findings demonstrate that NA-ergic modulation of the VTA-BLA circuit undergoes significant alterations in response to acute stress, with α2 -AR signaling indicated as a key target.


Asunto(s)
Transducción de Señal , Área Tegmental Ventral , Ratas , Animales , Masculino , Área Tegmental Ventral/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Transmisión Sináptica , Dopamina/metabolismo , Norepinefrina/metabolismo
2.
Eur J Neurosci ; 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36479859

RESUMEN

Drug seeking is associated with the ventral tegmental area (VTA) dopaminergic (DA) activity. Previously, we have shown that brief optogenetic inhibition of VTA DA neurons with 1 s pulses delivered every 9 s attenuates cocaine seeking under extinction conditions in rats without producing overt signs of dysphoria or locomotor sedation. Whether recruitment of neuronal pathways inhibiting VTA neuronal activity would suppress drug seeking remains unknown. Here, we asked if optogenetic stimulation of the lateral habenula (LHb) efferents in the rostromedial tegmental nucleus (RMTg) as well as RMTg efferents in VTA would reduce drug seeking. To investigate this, we measured how recruitment of elements of this inhibitory pathway affects cocaine seeking in male rats under extinction conditions. The effectiveness of brief optogenetic manipulations was confirmed electrophysiologically at the level of electrical activity of VTA DA neurons. Real-time conditioned place aversion (RT-CPA) and open field tests were performed to control for potential dysphoric/sedating effects of brief optogenetic stimulation of LHb-RMTg-VTA circuitry. Optogenetic stimulation of either RMTg or LHb inhibited VTA DAergic neuron firing, whereas similar stimulation of RMTg efferents in VTA or LHb efferents in RMTg reduced cocaine seeking under extinction conditions. Moreover, stimulation of LHb-RMTg efferents produced an effect that was maintained 24 h later, during cocaine seeking test without stimulation. This effect was specific, as brief optogenetic stimulation did not affect locomotor activity and was not aversive. Our results indicate that defined inhibitory pathways can be recruited to inhibit cocaine seeking, providing potential new targets for non-pharmacological treatment of drug craving.

3.
Cell Mol Neurobiol ; 42(7): 2423-2426, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33821329

RESUMEN

The development of tolerance and drug dependence limit the clinical application of opioids for the treatment of severe pain. Glucocorticoid receptors (GRs) are among molecular substrates involved in these processes. Most studies focus on the role of neuronal GR, while the involvement of GR on glial cells is not fully understood. To address this issue, we used a transgenic model of conditional GR knockout mice, targeted to connexin 30-expressing astrocytes, treated with repeated doses of morphine. We observed no difference between control mice and astrocytic GR knockouts in the development of antinociceptive tolerance. Nevertheless, when animals were subjected to precipitated withdrawal, knockouts presented some attenuated symptoms, including jumping. Taken together, our data suggest that hippocampal and spinal astrocytic GRs appear to be involved in opioid withdrawal, and drugs targeting the GR may relieve some symptoms of morphine withdrawal without influencing its antinociceptive properties.


Asunto(s)
Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Analgésicos Opioides , Animales , Astrocitos , Ratones , Ratones Noqueados , Morfina , Receptores de Glucocorticoides
4.
Psychol Health Med ; 27(5): 1084-1094, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33320724

RESUMEN

This study evaluated the frequency and risk factors for surgery dissatisfaction in patients undergoing lumbar or cervical surgery for degenerative spinal conditions. Based on the Patient Satisfaction Index (PSI) at 6 months after surgery, we divided patients into two groups: a satisfied and a dissatisfied group. We evaluated the association between patient dissatisfaction and five categories of variables:1) sociodemographic; 2) preoperative pain and disability [pain duration, level of surgery, previous spinal surgeries, pain scores as measured by the Short Form McGill Pain Questionnaire (SF-MPQ), numerical rating of average pain (NRS), disability as measured by the Oswestry Disability Index (ODI)]; 3) preoperative psychological status [depression, anxiety, and overall distress as measured by the Hospital Anxiety and Depression Scale (HADS), life satisfaction as measured by the Satisfaction With Life Scale (SWLS), and surgery expectations (SE) as measured by a Likert scale]; 4) postoperative improvements in pain and disability [improvements in SF-MPQ, improvement in ODI] and 5) postoperative psychological status [HADS, SWLS]. Results showed that 17.8% patients were dissatisfied with surgery. In the multivariate logistic analysis, more negative surgery expectations, smaller improvement in ODI scores, and a greater postoperative overall distress were significant risk factors associated with patient dissatisfaction with surgery.


Asunto(s)
Vértebras Lumbares , Satisfacción del Paciente , Evaluación de la Discapacidad , Humanos , Vértebras Lumbares/cirugía , Dolor , Factores de Riesgo , Resultado del Tratamiento
5.
Addict Biol ; 25(6): e12826, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31478293

RESUMEN

Preclinical studies strongly suggest that cocaine seeking depends on the neuronal activity of the ventral tegmental area (VTA) and phasic dopaminergic (DA) signaling. Notably, VTA pharmacological inactivation or dopamine receptor blockade in the forebrain may induce behavioral inhibition in general and acute aversive states in particular, thus reducing cocaine seeking indirectly. Such artifacts hinder successful translation of these findings in clinical studies and practice. Here, we aimed to evaluate if dynamic VTA manipulations effectively reduce cocaine seeking. We used male tyrosine hydroxylase (TH) IRES-Cre+ rats along with optogenetic tools to inhibit directly and briefly VTA DA neurons during conditioned stimulus (CS)-induced cocaine seeking under extinction conditions. The behavioral effects of optogenetic inhibition were also assessed in the real-time dynamic place aversion, conditioned place aversion, and CS-induced food-seeking tests. We found that brief and nondysphoric/nonsedative pulses of VTA photo-inhibition (1 s every 9 s, ie, for 10% of time) attenuated CS-induced cocaine seeking under extinction conditions in rats expressing archaerhodopsin selectively on the TH+ neurons. Furthermore, direct inhibition of the VTA DA activity reduced CS-induced cocaine seeking 24 hours after photo-modulation. Importantly, such effect appears to be selective for cocaine seeking as similar inhibition of the VTA DA activity had no effect on CS-induced food seeking. Thus, briefly inhibiting VTA DA activity during CS-induced cocaine seeking drastically and selectively reduces seeking without behavioral artifacts such as sedation or dysphoria. Our results point to the therapeutic possibilities of coupling nonpharmacologic treatments with extinction training in reducing cocaine addiction.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Trastornos Relacionados con Cocaína/psicología , Neuronas Dopaminérgicas/fisiología , Comportamiento de Búsqueda de Drogas/fisiología , Área Tegmental Ventral/fisiopatología , Animales , Cocaína/toxicidad , Condicionamiento Operante , Extinción Psicológica , Masculino , Inhibición Neural , Optogenética , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/genética
6.
Molecules ; 25(20)2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053718

RESUMEN

Molecular modeling approaches are an indispensable part of the drug design process. They not only support the process of searching for new ligands of a given receptor, but they also play an important role in explaining particular activity pathways of a compound. In this study, a comprehensive molecular modeling protocol was developed to explain the observed activity profiles of selected µ opioid receptor agents: two G protein-biased µ opioid receptor agonists(PZM21 and SR-17018), unbiased morphine, and the ß-arrestin-2-biased agonist,fentanyl. The study involved docking and molecular dynamics simulations carried out for three crystal structures of the target at a microsecond scale, followed by the statistical analysis of ligand-protein contacts. The interaction frequency between the modeled compounds and the subsequent residues of a protein during the simulation was also correlated with the output of in vitro and in vivo tests, resulting in the set of amino acids with the highest Pearson correlation coefficient values. Such indicated positions may serve as a guide for designing new G protein-biased ligands of the µ opioid receptor.


Asunto(s)
Morfina/química , Receptores Opioides/metabolismo , Animales , Fentanilo/química , Fentanilo/metabolismo , Humanos , Simulación de Dinámica Molecular , Receptores Opioides/química , Tiofenos/química , Urea/análogos & derivados , Urea/química
7.
Addict Biol ; 23(2): 596-609, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28635140

RESUMEN

Exposure to drug-associated cues evokes drug-seeking behavior and is regarded as a major cause of relapse. Conditional stimulus upregulates noradrenaline (NA) system activity, but the drug-seeking behavior depends particularly on phasic dopamine signaling downstream from the ventral tegmental area (VTA). The VTA dopamine-ergic activity is regulated via the signaling of alpha1 -adrenergic and alpha2 -adrenergic receptors (α1 -ARs and α2 -ARs); thus, the impact of the conditional stimulus on drug-seeking behavior might involve NAergic signaling in the VTA. To date, the role of VTA ARs in regulating cocaine seeking was not studied. We found that cocaine seeking under extinction conditions in male Sprague-Dawley rats was attenuated by intra-VTA prazosin or terazosin-two selective α1 -AR antagonists. In contrast, cocaine seeking was facilitated by intra-VTA administration of the selective α1 -AR agonist phenylephrine as well as α2 -AR antagonist RX 821002, whereas the selective ß-AR antagonist propranolol had no effects. In addition, blockade of α1 -AR in the VTA prevented α2 -AR antagonist-induced enhancement of cocaine seeking. Importantly, the potential non-specific effects of the VTA AR blockade on cocaine seeking could be excluded, because none of the AR antagonists influenced sucrose seeking under extinction conditions or locomotor activity in the open field test. These results demonstrate that NAergic signaling potently and selectively regulates cocaine seeking during early cocaine withdrawal via VTA α1 -AR and α2 -AR but not ß-AR. Our findings provide new insight into the NAergic mechanisms that underlie cocaine craving.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Conducta Animal/efectos de los fármacos , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Área Tegmental Ventral , Antagonistas Adrenérgicos beta/farmacología , Animales , Condicionamiento Operante , Ansia , Idazoxan/análogos & derivados , Idazoxan/farmacología , Masculino , Fenilefrina/farmacología , Prazosina/análogos & derivados , Prazosina/farmacología , Propranolol/farmacología , Ratas , Ratas Sprague-Dawley , Autoadministración
8.
BMC Neurosci ; 18(1): 37, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28381250

RESUMEN

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.


Asunto(s)
Astrocitos/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores de Esteroides/agonistas , Esteroides/farmacología , Transcriptoma/efectos de los fármacos , Animales , Astrocitos/metabolismo , Células Cultivadas , Análisis por Conglomerados , Cuerpo Estriado/metabolismo , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Análisis por Micromatrices , Neuronas/metabolismo , ARN Mensajero/metabolismo , Receptores de Esteroides/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma/fisiología
9.
Nature ; 473(7347): 372-5, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21508957

RESUMEN

A minority of individuals experiencing traumatic events develop anxiety disorders. The reason for the lack of correspondence between the prevalence of exposure to psychological trauma and the development of anxiety is unknown. Extracellular proteolysis contributes to fear-associated responses by facilitating neuronal plasticity at the neuron-matrix interface. Here we show in mice that the serine protease neuropsin is critical for stress-related plasticity in the amygdala by regulating the dynamics of the EphB2-NMDA-receptor interaction, the expression of Fkbp5 and anxiety-like behaviour. Stress results in neuropsin-dependent cleavage of EphB2 in the amygdala causing dissociation of EphB2 from the NR1 subunit of the NMDA receptor and promoting membrane turnover of EphB2 receptors. Dynamic EphB2-NR1 interaction enhances NMDA receptor current, induces Fkbp5 gene expression and enhances behavioural signatures of anxiety. On stress, neuropsin-deficient mice do not show EphB2 cleavage and its dissociation from NR1 resulting in a static EphB2-NR1 interaction, attenuated induction of the Fkbp5 gene and low anxiety. The behavioural response to stress can be restored by intra-amygdala injection of neuropsin into neuropsin-deficient mice and disrupted by the injection of either anti-EphB2 antibodies or silencing the Fkbp5 gene in the amygdala of wild-type mice. Our findings establish a novel neuronal pathway linking stress-induced proteolysis of EphB2 in the amygdala to anxiety.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Calicreínas/metabolismo , Receptor EphB2/metabolismo , Amígdala del Cerebelo/citología , Animales , Ansiedad/genética , Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/genética , Trastornos de Ansiedad/metabolismo , Conductividad Eléctrica , Miedo , Regulación de la Expresión Génica , Calicreínas/deficiencia , Calicreínas/genética , Potenciación a Largo Plazo , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Neuronas/metabolismo , Unión Proteica , Receptor EphB2/química , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Estrés Psicológico/metabolismo , Proteínas de Unión a Tacrolimus/genética
10.
Addict Biol ; 22(6): 1802-1816, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27578564

RESUMEN

Chronic exposure to opioids induces adaptations in brain function that lead to the formation of the behavioral and physiological symptoms of drug dependence and addiction. Animal models commonly used to test these symptoms typically last less than two weeks, which is presumably too short to observe the alterations in the brain that accompany drug addiction. Here, we analyzed the phenotypic and molecular effects of nearly lifelong morphine or saccharin intake in C57BL/6J mice. We used multiple paradigms to evaluate the symptoms of compulsive drug intake: a progressive ratio schedule, intermittent access and a schedule involving a risk of punishment were programmed into an automated IntelliCage system. Gene expression profiles were evaluated in the striatum using whole-genome microarrays and further validated using quantitative polymerase chain reaction in the striatum and the prefrontal cortex. Mice voluntary self-administering morphine showed addiction-related behavioral pattern that included: higher motivation to work for a drug reward, increased reward seeking and increased craving. The analysis of molecular changes revealed a tolerance effect in the transcriptional response to morphine injection (20 mg/kg, ip), as well as some long-lasting alterations in gene expression profiles between the analyzed groups of animals. Interestingly, among the morphine-drinking animals, certain transcriptional profiles were found to be associated with alterations in behavior. In conclusion, our model represents a novel approach for investigating the behavioral and molecular mechanisms underlying opioid addiction. Prolonged morphine intake caused adaptive processes in the brain that manifested as altered behavior and transcriptional sensitivity to opioids.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Trastornos Relacionados con Opioides/fisiopatología , Analgésicos Opioides/administración & dosificación , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Autoadministración
11.
BMC Bioinformatics ; 17: 85, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26868127

RESUMEN

BACKGROUND: The regulation of gene expression in eukaryotic cells is a complex process that involves epigenetic modifications and the interaction of DNA with multiple transcription factors. This process can be studied with unprecedented sensitivity using a combination of chromatin immunoprecipitation and next-generation DNA sequencing (ChIP-seq). Available ChIP-seq data can be further utilized to interpret new gene expression profiling experiments. RESULTS: Here, we describe seqinspector, a tool that accepts any set of genomic coordinates from ChIP-seq or RNA-seq studies to identify shared transcriptional regulators. The presented web resource includes a large collection of publicly available ChIP-seq and RNA-seq experiments (>1300 tracks) performed on transcription factors, histone modifications, RNA polymerases, enhancers and insulators in humans and mice. Over-representation is calculated based on the coverage computed directly from indexed files storing ChIP-seq data (bigwig). Therefore, seqinspector is not limited to pre-computed sets of gene promoters. CONCLUSION: The tool can be used to identify common gene expression regulators for sets of co-expressed transcripts (including miRNAs, lncRNAs or any novel unannotated RNAs) or for sets of ChIP-seq peaks to identify putative protein-protein interactions or transcriptional co-factors. The tool is available at http://seqinspector.cremag.org.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Transcriptoma , Animales , Epigénesis Genética/genética , Genómica/métodos , Humanos , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , Factores de Transcripción/metabolismo
12.
BMC Genomics ; 17: 362, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27188165

RESUMEN

BACKGROUND: The NMDA receptor antagonist ketamine was found to act as a fast-acting antidepressant. The effects of single treatment were reported to persist for days to weeks, even in otherwise treatment-refractory cases. Identification of the mechanisms underlying ketamine's antidepressant action may permit development of novel drugs, with similar clinical properties but lacking psychotomimetic, sedative and other side effects. METHODS: We applied whole-genome microarray profiling to analyze detailed time-course (1, 2, 4 and 8 h) of transcriptome alterations in the striatum and hippocampus following acute administration of ketamine, memantine and phencyclidine in C57BL/6 J mice. The transcriptional effects of ketamine were further analyzed using next-generation sequencing and quantitative PCR. Gene expression alterations induced by the NMDA antagonists were compared to the molecular profiles of psychotropic drugs: antidepressants, antipsychotics, anxiolytics, psychostimulants and opioids. RESULTS: We identified 52 transcripts (e.g. Dusp1, Per1 and Fkbp5) with altered expression (FDR < 1 %) in response to treatment with NMDA receptor antagonists. Functional links that connect expression of the regulated genes to the MAPK, IL-6 and insulin signaling pathways were indicated. Moreover, ketamine-regulated expression of specific gene isoforms was detected (e.g. Tsc22d3, Sgk1 and Hif3a). The comparison with other psychotropic drugs revealed that the molecular effects of ketamine are most similar to memantine and phencyclidine. Clustering based on expression profiles placed the NMDA antagonists among fluoxetine, tianeptine, as well as opioids and ethanol. CONCLUSIONS: The identified patterns of gene expression alteration in the brain provided novel molecular classification of ketamine. The transcriptional profile of ketamine reflects its multi-target pharmacological nature. The results reveal similarities between the effects of ketamine and monoaminergic antidepressants that may explain the mechanisms of its rapid antidepressant action.


Asunto(s)
Anestésicos Disociativos/farmacología , Antidepresivos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ketamina/farmacología , Transcriptoma , Animales , Análisis por Conglomerados , Biología Computacional/métodos , Cuerpo Estriado/metabolismo , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Hipocampo/metabolismo , Ligandos , Masculino , Ratones , Receptores de N-Metil-D-Aspartato/genética , Reproducibilidad de los Resultados
13.
Przegl Lek ; 73(9): 648-51, 2016.
Artículo en Polaco | MEDLINE | ID: mdl-29688673

RESUMEN

Introduction: Cerebral stroke can lead both to the limitation of motor skills and cognitive dysfunctions as well as mood disorders. One of the most frequent results of cerebral stroke is post-stroke depression (PSD). Numerous researchers have emphasized the significance of an early detection of this disorder. There is discussion concerning both its background and the methods that might be used for its evaluation. One of the scales applied for this purpose is Post Stroke Depression Rating Scale (PSDRS). Material and methods: The aim of this study was to determine the profile of depressive symptoms in patients at an early stage after stroke. The participants of the study were 43 persons who underwent cerebral stroke. The study was carried out in the course of the first week after the vascular incident. The patients were examined with the application of the PSDRS elaborated in order to determine the specific nature of post-stroke depression. Results: The results of the PSDRS applied indicate the occurrence of three factors present in the group under study. The first factor includes the symptoms related to a depressive mood, sense of guilt, apathy and suicidal thoughts; the second one is related to the impairment of emotional control, anhedonia, variation over time and a catastrophic reaction; the third factor concerns vegetative disorders and anxiety. Conclusions: The application of the PSDRS for the evaluation of depressed mood in persons in the acute phase after cerebral stroke enables the distinction of the factors covering various aspects related to post-stroke depression. This scale is a valuable tool for the evaluation of early symptoms of depression in patients in the first week after cerebral stroke.


Asunto(s)
Depresión/diagnóstico , Escalas de Valoración Psiquiátrica , Accidente Cerebrovascular/psicología , Adulto , Anciano , Anciano de 80 o más Años , Humanos , Persona de Mediana Edad
14.
Mol Pain ; 10: 47, 2014 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-25038616

RESUMEN

BACKGROUND: The molecular mechanisms underlying neuropathic pain are constantly being studied to create new opportunities to prevent or alleviate neuropathic pain. The aim of our study was to determine the gene expression changes induced by sciatic nerve chronic constriction injury (CCI) that are modulated by minocycline, which can effectively diminish neuropathic pain in animal studies. The genes associated with minocycline efficacy in neuropathic pain should provide insight into the etiology of neuropathic pain and identify novel therapeutic targets. RESULTS: We screened the ipsilateral dorsal part of the lumbar spinal cord of the rat CCI model for differentially expressed genes. Out of 22,500 studied transcripts, the abundance levels of 93 transcripts were altered following sciatic nerve ligation. Percentage analysis revealed that 54 transcripts were not affected by the repeated administration of minocycline (30 mg/kg, i.p.), but the levels of 39 transcripts were modulated following minocycline treatment. We then selected two gene expression patterns, B1 and B2. The first transcription pattern, B1, consisted of 10 mRNA transcripts that increased in abundance after injury, and minocycline treatment reversed or inhibited the effect of the injury; the B2 transcription pattern consisted of 7 mRNA transcripts whose abundance decreased following sciatic nerve ligation, and minocycline treatment reversed the effect of the injury. Based on the literature, we selected seven genes for further analysis: Cd40, Clec7a, Apobec3b, Slc7a7, and Fam22f from pattern B1 and Rwdd3 and Gimap5 from pattern B2. Additionally, these genes were analyzed using quantitative PCR to determine the transcriptional changes strongly related to the development of neuropathic pain; the ipsilateral DRGs (L4-L6) were also collected and analyzed in these rats using qPCR. CONCLUSION: In this work, we confirmed gene expression alterations previously identified by microarray analysis in the spinal cord and analyzed the expression of selected genes in the DRG. Moreover, we reviewed the literature to illustrate the relevance of these findings for neuropathic pain development and therapy. Further studies are needed to elucidate the roles of the individual genes in neuropathic pain and to determine the therapeutic role of minocycline in the rat neuropathic pain model.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Minociclina/farmacología , Ciática/metabolismo , Sistema de Transporte de Aminoácidos y+ , Analgésicos no Narcóticos/uso terapéutico , Animales , Antígenos CD40 , Citidina Desaminasa , Modelos Animales de Enfermedad , Lateralidad Funcional , Perfilación de la Expresión Génica , Lectinas Tipo C , Masculino , Glicoproteínas de Membrana , Minociclina/uso terapéutico , Análisis de Secuencia por Matrices de Oligonucleótidos , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Wistar , Ciática/tratamiento farmacológico , Ciática/patología , Médula Espinal/metabolismo
15.
Addict Biol ; 19(4): 643-51, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23301597

RESUMEN

Experimental evidence suggests that endogenous opioids play an important role in the development of ethanol addiction. In this study, we employed two mouse lines divergently bred for opioid-mediated stress-induced analgesia. In comparison with HA (high analgesia line) mice, LA (low analgesia line) mice, having lower opioid receptor system activity, manifest enhanced basal as well as stress-induced ethanol drinking. Here, we found that recently discovered C320T transition in exon 2 of the δ-opioid receptor gene (EU446125.1), which results in an A107V substitution (ACA23171.1), leads to higher ethanol preference in CT mice compared with CC homozygotes. This genetic association is particularly evident under chronic mild stress (CMS) conditions. The interaction between stress and ethanol intake was significantly stronger in HA than in LA mice. Ethanol almost completely attenuated the pro-depressive effect of CMS (assessed with the tail suspension test) in both the CC and CT genotypes in the HA line. In the LA mice, a lack of response to ethanol was observed in the CC genotype, whereas ethanol consumption strengthened depressive-like behaviours in CT individuals. Our results suggest that constitutively active A107V substitution in δ-opioid receptors may be involved in stress-enhanced vulnerability to ethanol abuse and in the risk of ethanol dependence.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Péptidos Opioides/genética , Polimorfismo Genético/genética , Receptores Opioides delta/genética , Estrés Psicológico/fisiopatología , Analgesia , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Ratones , Ratones Endogámicos , Péptidos Opioides/fisiología
16.
Glia ; 61(4): 623-35, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23339081

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Marcación de Gen/métodos , Morfina/administración & dosificación , Neuronas/fisiología , Receptores de Glucocorticoides/fisiología , Transducción de Señal/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
17.
BMC Genomics ; 14: 606, 2013 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-24010892

RESUMEN

BACKGROUND: Despite their widespread use, the biological mechanisms underlying the efficacy of psychotropic drugs are still incompletely known; improved understanding of these is essential for development of novel more effective drugs and rational design of therapy. Given the large number of psychotropic drugs available and their differential pharmacological effects, it would be important to establish specific predictors of response to various classes of drugs. RESULTS: To identify the molecular mechanisms that may initiate therapeutic effects, whole-genome expression profiling (using 324 Illumina Mouse WG-6 microarrays) of drug-induced alterations in the mouse brain was undertaken, with a focus on the time-course (1, 2, 4 and 8 h) of gene expression changes produced by eighteen major psychotropic drugs: antidepressants, antipsychotics, anxiolytics, psychostimulants and opioids. The resulting database is freely accessible at http://www.genes2mind.org. Bioinformatics approaches led to the identification of three main drug-responsive genomic networks and indicated neurobiological pathways that mediate the alterations in transcription. Each tested psychotropic drug was characterized by a unique gene network expression profile related to its neuropharmacological properties. Functional links that connect expression of the networks to the development of neuronal adaptations (MAPK signaling pathway), control of brain metabolism (adipocytokine pathway), and organization of cell projections (mTOR pathway) were found. CONCLUSIONS: The comparison of gene expression alterations between various drugs opened a new means to classify the different psychoactive compounds and to predict their cellular targets; this is well exemplified in the case of tianeptine, an antidepressant with unknown mechanisms of action. This work represents the first proof-of-concept study of a molecular classification of psychoactive drugs.


Asunto(s)
Encéfalo/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Psicotrópicos/farmacología , Animales , Biología Computacional , Bases de Datos Genéticas , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcriptoma
18.
Behav Brain Res ; 449: 114466, 2023 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-37146718

RESUMEN

A growing body of evidence points out the involvement of the µ-opioid receptors in the modulation of stress-related behaviour. It has been suggested that µ-opioid receptor agonists may attenuate behavioural despair following animals' exposure to an acute, inescapable stressor. Moreover, morphine was shown to ameliorate fear memories caused by a traumatic experience. As typical µ-opioid receptor agonists entail a risk of serious side effects and addiction, novel, possibly safer and less addictive agonists of this receptor are currently under investigation. One of them, PZM21, preferentially acting via the G protein signalling pathway, was previously shown to be analgesic, but less addictive than morphine. Here, we aimed to further test this ligand in stress-related behavioural paradigms in mice. The study has shown that, unlike morphine, PZM21 does not decrease immobility in the forced swimming and tail suspension tests. On the other hand, we observed that both mice treated with PZM21 and those receiving morphine presented a slight attenuation of freezing across the consecutive fear memory retrievals in the fear conditioning test. Therefore, our study implies that at the range of tested doses, PZM21, a nonrewarding representative of G protein-biased µ-opioid receptor agonists, may interfere with fear memory consolidation while having no beneficial effects on behavioural despair in mice.


Asunto(s)
Analgésicos Opioides , Receptores Opioides mu , Ratones , Animales , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacología , Morfina/farmacología , Proteínas de Unión al GTP/metabolismo , Receptores Opioides/metabolismo , Miedo
19.
Nat Commun ; 14(1): 2134, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37185241

RESUMEN

Severe psychological trauma triggers genetic, biochemical and morphological changes in amygdala neurons, which underpin the development of stress-induced behavioural abnormalities, such as high levels of anxiety. miRNAs are small, non-coding RNA fragments that orchestrate complex neuronal responses by simultaneous transcriptional/translational repression of multiple target genes. Here we show that miR-483-5p in the amygdala of male mice counterbalances the structural, functional and behavioural consequences of stress to promote a reduction in anxiety-like behaviour. Upon stress, miR-483-5p is upregulated in the synaptic compartment of amygdala neurons and directly represses three stress-associated genes: Pgap2, Gpx3 and Macf1. Upregulation of miR-483-5p leads to selective contraction of distal parts of the dendritic arbour and conversion of immature filopodia into mature, mushroom-like dendritic spines. Consistent with its role in reducing the stress response, upregulation of miR-483-5p in the basolateral amygdala produces a reduction in anxiety-like behaviour. Stress-induced neuromorphological and behavioural effects of miR-483-5p can be recapitulated by shRNA mediated suppression of Pgap2 and prevented by simultaneous overexpression of miR-483-5p-resistant Pgap2. Our results demonstrate that miR-483-5p is sufficient to confer a reduction in anxiety-like behaviour and point to miR-483-5p-mediated repression of Pgap2 as a critical cellular event offsetting the functional and behavioural consequences of psychological stress.


Asunto(s)
Complejo Nuclear Basolateral , MicroARNs , Animales , Masculino , Ratones , Amígdala del Cerebelo/metabolismo , Complejo Nuclear Basolateral/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo
20.
Int J Neuropsychopharmacol ; 15(10): 1457-71, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22040728

RESUMEN

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.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Proteínas Portadoras/biosíntesis , Ácido Glutámico/fisiología , Dependencia de Morfina/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Desempeño Psicomotor/fisiología , Neuronas Adrenérgicas/efectos de los fármacos , Antagonistas Adrenérgicos alfa/farmacología , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dependencia de Morfina/genética , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Técnicas de Cultivo de Órganos , Desempeño Psicomotor/efectos de los fármacos , Receptores de N-Metil-D-Aspartato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA