Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Epigenetics ; 10(3): 247-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793778

RESUMEN

Early-life stress (ELS) induces long-lasting changes in gene expression conferring an increased risk for the development of stress-related mental disorders. Glucocorticoid receptors (GR) mediate the negative feedback actions of glucocorticoids (GC) in the paraventricular nucleus (PVN) of the hypothalamus and anterior pituitary and therefore play a key role in the regulation of the hypothalamic-pituitary-adrenal (HPA) axis and the endocrine response to stress. We here show that ELS programs the expression of the GR gene (Nr3c1) by site-specific hypermethylation at the CpG island (CGI) shore in hypothalamic neurons that produce corticotropin-releasing hormone (Crh), thus preventing Crh upregulation under conditions of chronic stress. CpGs mapping to the Nr3c1 CGI shore region are dynamically regulated by ELS and underpin methylation-sensitive control of this region's insulation-like function via Ying Yang 1 (YY1) binding. Our results provide new insight into how a genomic element integrates experience-dependent epigenetic programming of the composite proximal Nr3c1 promoter, and assigns an insulating role to the CGI shore.


Asunto(s)
Metilación de ADN , Regiones Promotoras Genéticas , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/metabolismo , Animales , Células Cultivadas , Islas de CpG , Hipotálamo/metabolismo , Masculino , Ratones Endogámicos C57BL
3.
BMC Genomics ; 13: 579, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23114097

RESUMEN

BACKGROUND: The hypothalamic-pituitary-adrenal (HPA) axis is essential to control physiological stress responses in mammals. Its dysfunction is related to several mental disorders, including anxiety and depression. The aim of this study was to identify genetic loci underlying the endocrine regulation of the HPA axis. METHOD: High (HAB) and low (LAB) anxiety-related behaviour mice were established by selective inbreeding of outbred CD-1 mice to model extremes in trait anxiety. Additionally, HAB vs. LAB mice exhibit comorbid characteristics including a differential corticosterone response upon stress exposure. We crossbred HAB and LAB lines to create F1 and F2 offspring. To identify the contribution of the endocrine phenotypes to the total phenotypic variance, we examined multiple behavioural paradigms together with corticosterone secretion-based phenotypes in F2 mice by principal component analysis. Further, to pinpoint the genomic loci of the quantitative trait of the HPA axis stress response, we conducted genome-wide multipoint oligogenic linkage analyses based on Bayesian Markov chain Monte Carlo approach as well as parametric linkage in three-generation pedigrees, followed by a two-dimensional scan for epistasis and association analysis in freely segregating F2 mice using 267 single-nucleotide polymorphisms (SNPs), which were identified to consistently differ between HAB and LAB mice as genetic markers. RESULTS: HPA axis reactivity measurements and behavioural phenotypes were represented by independent principal components and demonstrated no correlation. Based on this finding, we identified one single quantitative trait locus (QTL) on chromosome 3 showing a very strong evidence for linkage (2ln (L-score) > 10, LOD > 23) and significant association (lowest Bonferroni adjusted p < 10-28) to the neuroendocrine stress response. The location of the linkage peak was estimated at 42.3 cM (95% confidence interval: 41.3 - 43.3 cM) and was shown to be in epistasis (p-adjusted < 0.004) with the locus at 35.3 cM on the same chromosome. The QTL harbours genes involved in steroid synthesis and cardiovascular effects. CONCLUSION: The very prominent effect on stress-induced corticosterone secretion of the genomic locus on chromosome 3 and its involvement in epistasis highlights the critical role of this specific locus in the regulation of the HPA axis.


Asunto(s)
Ansiedad/genética , Ansiedad/fisiopatología , Cromosomas de los Mamíferos/genética , Sistema Endocrino/fisiología , Sitios de Carácter Cuantitativo/genética , Estrés Fisiológico/genética , Glándulas Suprarrenales/fisiopatología , Animales , Sistema Endocrino/metabolismo , Femenino , Marcadores Genéticos/genética , Hipotálamo/fisiopatología , Masculino , Ratones , Fenotipo , Hipófisis/fisiopatología
4.
J Psychiatr Res ; 45(5): 650-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21106206

RESUMEN

Some, but not all studies in patients with posttraumatic stress disorder (PTSD), report reduced hippocampus (HPC) volume. In particular it is unclear, whether smaller hippocampal volume represents a susceptibility factor for PTSD rather than a consequence of the trauma. To gain insight into the relationship of brain morphology and trauma exposure, we investigated volumetric and molecular changes of the HPC in a mouse model of PTSD by means of in vivo Manganese Enhanced Magnetic Resonance Imaging (MEMRI) and ex vivo ultramicroscopic measurements. Exposure to a brief inescapable foot shock led to a volume reduction in both left HPC and right central amygdala two months later. This volume loss was mirrored by a down-regulation of growth-associated protein-43 (GAP43) in the HPC. Enriched housing decreased the intensity of trauma-associated contextual fear, independently of whether it was provided before or after the shock. Beyond that, enriched housing led to an increase in intracranial volume, including the lateral ventricles and the hippocampus, and to an up-regulation of GAP43 as revealed by MEMRI and Western blot analysis, thus partially compensating for trauma-related HPC volume loss and down-regulation of GAP43 expression. Together these data demonstrate that traumatic experience in mice causes a reduction in HPC and central amygdala volume possibly due to a shrinkage of axonal protrusions. Enriched housing might induce trophic changes, which may contribute to the amelioration of trauma-associated PTSD-like symptoms at behavioural, morphological and molecular levels.


Asunto(s)
Hipocampo/patología , Trastornos por Estrés Postraumático/patología , Estimulación Acústica , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ambiente , Miedo/fisiología , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica/fisiología , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética/métodos , Masculino , Manganeso , Ratones , Ratones Endogámicos C57BL , Reflejo/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/rehabilitación , Factores de Tiempo
5.
J Neurosci ; 30(27): 9103-16, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20610744

RESUMEN

Urocortin 3 (UCN3) is strongly expressed in specific nuclei of the rodent brain, at sites distinct from those expressing urocortin 1 and urocortin 2, the other endogenous ligands of corticotropin-releasing hormone receptor type 2 (CRH-R2). To determine the physiological role of UCN3, we generated UCN3-deficient mice, in which the UCN3 open reading frame was replaced by a tau-lacZ reporter gene. By means of this reporter gene, the nucleus parabrachialis and the premammillary nucleus were identified as previously unknown sites of UCN3 expression. Additionally, the introduced reporter gene enabled the visualization of axonal projections of UCN3-expressing neurons from the superior paraolivary nucleus to the inferior colliculus and from the posterodorsal part of the medial amygdala to the principal nucleus of the bed nucleus of the stria terminalis, respectively. The examination of tau-lacZ reporter gene activity throughout the brain underscored a predominant expression of UCN3 in nuclei functionally connected to the accessory olfactory system. Male and female mice were comprehensively phenotyped but none of the applied tests provided indications for a role of UCN3 in the context of hypothalamic-pituitary-adrenocortical axis regulation, anxiety- or depression-related behavior. However, inspired by the prevalent expression throughout the accessory olfactory system, we identified alterations in social discrimination abilities of male and female UCN3 knock-out mice that were also present in male CRH-R2 knock-out mice. In conclusion, our results suggest a novel role for UCN3 and CRH-R2 related to the processing of social cues and to the establishment of social memories.


Asunto(s)
Discriminación en Psicología/fisiología , Relaciones Interpersonales , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Reconocimiento en Psicología/fisiología , Urocortinas/metabolismo , Estimulación Acústica/métodos , Animales , Encéfalo/citología , Encéfalo/metabolismo , Ritmo Circadiano/fisiología , Corticosterona/sangre , Miedo/fisiología , Femenino , Regulación de la Expresión Génica/genética , Sistema Hipotálamo-Hipofisario/metabolismo , Inhibición Psicológica , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Odorantes , Vías Olfatorias/fisiología , Percepción/fisiología , Sistema Hipófiso-Suprarrenal/embriología , Radioinmunoensayo/métodos , Receptores de Hormona Liberadora de Corticotropina/deficiencia , Reflejo de Sobresalto/genética , Estadísticas no Paramétricas , Natación/fisiología , Urocortinas/deficiencia
6.
Eur Neuropsychopharmacol ; 20(5): 346-55, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20097046

RESUMEN

Due to strict exclusion criteria the generalizability of randomized controlled trials appears to be limited. Therefore, outcomes of naturalistically treated depressive inpatients with respect to depression mean scores, response and remission rates were evaluated. This was a multicenter trial, conducted in 12 psychiatric hospitals in Germany with a follow-up period of 4years. Patients were assessed biweekly from admission to discharge with diverse psychopathological rating scales. All patients (n=1014) met DSM-IV criteria for major depressive episode. Results are presented only for the acute inpatient treatment period. Mean inpatient treatment duration was 53.6+/-47.5days. Reduction on depression scales was evident as soon as week 2 and remained significant. Mean HAMD-17 total score decreased from 22.3 to 8.8. A total of 68.9% were classified as responders (> or =50% reduction of the initial HAMD-17 score), whereas 51.9% achieved remission (HAMD-17 total score < or =7). Of those who ultimately achieved response more than 40% did so within the first 2weeks. An individualized naturalistic inpatient treatment approach appears to be beneficial in terms of effectiveness.


Asunto(s)
Trastorno Depresivo Mayor/terapia , Adolescente , Adulto , Anciano , Antidepresivos/uso terapéutico , Terapia Electroconvulsiva , Femenino , Estudios de Seguimiento , Humanos , Pacientes Internos , Masculino , Persona de Mediana Edad , Selección de Paciente , Fototerapia , Escalas de Valoración Psiquiátrica , Índice de Severidad de la Enfermedad , Privación de Sueño , Estimulación Magnética Transcraneal , Resultado del Tratamiento
7.
PLoS One ; 4(8): e6749, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19707599

RESUMEN

BACKGROUND: A condition vital for the consolidation and maintenance of sleep is generally reduced responsiveness to external stimuli. Despite this, the sleeper maintains a level of stimulus processing that allows to respond to potentially dangerous environmental signals. The mechanisms that subserve these contradictory functions are only incompletely understood. METHODOLOGY/PRINCIPAL FINDINGS: Using combined EEG/fMRI we investigated the neural substrate of sleep protection by applying an acoustic oddball paradigm during light NREM sleep. Further, we studied the role of evoked K-complexes (KCs), an electroencephalographic hallmark of NREM sleep with a still unknown role for sleep protection. Our main results were: (1) Other than in wakefulness, rare tones did not induce a blood oxygenation level dependent (BOLD) signal increase in the auditory pathway but a strong negative BOLD response in motor areas and the amygdala. (2) Stratification of rare tones by the presence of evoked KCs detected activation of the auditory cortex, hippocampus, superior and middle frontal gyri and posterior cingulate only for rare tones followed by a KC. (3) The typical high frontocentral EEG deflections of KCs were not paralleled by a BOLD equivalent. CONCLUSIONS/SIGNIFICANCE: We observed that rare tones lead to transient disengagement of motor and amygdala responses during light NREM sleep. We interpret this as a sleep protective mechanism to delimit motor responses and to reduce the sensitivity of the amygdala towards further incoming stimuli. Evoked KCs are suggested to originate from a brain state with relatively increased stimulus processing, revealing an activity pattern resembling novelty processing as previously reported during wakefulness. The KC itself is not reflected by increased metabolic demand in BOLD based imaging, arguing that evoked KCs result from increased neural synchronicity without altered metabolic demand.


Asunto(s)
Estimulación Acústica , Electroencefalografía , Imagen por Resonancia Magnética , Sueño/fisiología , Adulto , Potenciales Evocados , Femenino , Humanos , Masculino
8.
Eur J Neurosci ; 25(3): 863-71, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17328781

RESUMEN

High thalamocortical neuronal activity characterizes both, wakefulness and rapid eye movement (REM) sleep, but apparently this network fulfills other roles than processing external information during REM sleep. To investigate thalamic and cortical reactivity during human REM sleep, we used functional magnetic resonance imaging with simultaneous polysomnographic recordings while applying acoustic stimulation. Our observations indicate two distinct functional substates within general REM sleep. Acoustic stimulation elicited a residual activation of the auditory cortex during tonic REM sleep background without rapid eye movements. By contrast, periods containing bursts of phasic activity such as rapid eye movements appear characterized by a lack of reactivity to sensory stimuli. We report a thalamocortical network including limbic and parahippocampal areas specifically active during phasic REM periods. Thus, REM sleep has to be subdivided into tonic REM sleep with residual alertness, and phasic REM sleep with the brain acting as a functionally isolated and closed intrinsic loop.


Asunto(s)
Corteza Auditiva/fisiología , Imagen por Resonancia Magnética , Sueño REM/fisiología , Tálamo/fisiología , Estimulación Acústica , Adulto , Corteza Auditiva/citología , Femenino , Humanos , Masculino , Vías Nerviosas , Giro Parahipocampal/citología , Giro Parahipocampal/fisiología , Polisomnografía , Tálamo/citología
9.
Neuropsychopharmacology ; 32(9): 1941-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17287823

RESUMEN

There is an extensive evidence that corticotropin releasing factor (CRF) is hypersecreted in depression and anxiety, and blockade of CRF could have therapeutic benefit. We report preclinical data and the results of a clinical Phase I study with the novel nonpeptide CRF(1) antagonist NBI-34041/SB723620. Preclinical data conducted with different cell lines expressing human CRF receptors and in Wistar and Sprague-Dawley rats indicate that NBI-34041 is effective in reducing endocrine responses to pharmacological and behavioral challenge mediated by CRF(1) receptors. These specific properties and its well-documented safety profile enabled a clinical Phase I study with 24 healthy male subjects receiving NBI-34041 (10, 50, or 100 mg) or placebo for 14 days. Regulation of the hypothalamic-pituitary-adrenocortical (HPA) axis was evaluated by intravenous stimulation with 100 microg of human CRF. Psychosocial stress response was investigated with the Trier Social Stress Test (TSST). Treatment with NBI-34041 did not impair diurnal adrenocorticotropic hormone (ACTH) and cortisol secretion or CRF evoked ACTH and cortisol responses but attenuated the neuroendocrine response to psychosocial stress. These results suggest that NBI-34041 is safe and does not impair basal regulation of the HPA system but improves resistance against psychosocial stress. NBI-34041 demonstrates that inhibition of the CRF system is a promising target for drug development against depression and anxiety disorders.


Asunto(s)
Hormona Liberadora de Corticotropina/uso terapéutico , Evaluación Preclínica de Medicamentos , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/fisiología , Estrés Psicológico/tratamiento farmacológico , Acenaftenos/uso terapéutico , Hormona Adrenocorticotrópica/sangre , Análisis de Varianza , Animales , Autorradiografía , Línea Celular , Hormona Liberadora de Corticotropina/farmacología , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Emociones/efectos de los fármacos , Humanos , Hidrocortisona/sangre , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
10.
Neuroreport ; 16(8): 853-7, 2005 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-15891584

RESUMEN

In animal models, ponto-geniculo-occipital waves appear as an early sign of rapid eye movement sleep and may be functionally significant for brain plasticity processes. In this pilot study, we use a combined polysomnographic and functional magnetic resonance imaging approach, and show distinct magnetic resonance imaging signal increases in the posterior thalamus and occipital cortex in close temporal relationship to rapid eye movements during human rapid eye movement sleep. These findings are consistent with cell recordings in animal experiments and demonstrate that functional magnetic resonance imaging can be utilized to detect ponto-geniculo-occipital-like activity in humans. Studying intact neuronal networks underlying sleep regulation is no longer confined to animal models, but has been shown to be feasible in humans by a combined functional magnetic resonance imaging and electroencephalograph approach.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Lóbulo Occipital/irrigación sanguínea , Sueño REM/fisiología , Sueño/fisiología , Tálamo/irrigación sanguínea , Adulto , Mapeo Encefálico , Mentón/fisiología , Electroencefalografía/métodos , Electromiografía/métodos , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Masculino , Lóbulo Occipital/fisiología , Oxígeno/sangre , Proyectos Piloto , Polisomnografía/métodos , Tálamo/fisiología
11.
Proc Natl Acad Sci U S A ; 102(17): 6183-8, 2005 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-15833812

RESUMEN

Corticotropin-releasing hormone (CRH) coordinates hormonal and behavioral responses to stress. The mitogen-activated protein kinase extracellular signal-related kinase 1/2 (ERK1/2) mediates several functions in different forebrain structures and recently has been implicated in CRH signaling in cultured cells. To study in vivo CRH-mediated activation of central ERK1/2, we investigated the expression pattern of the phosphorylated ERK1/2(p-ERK1/2) in the mouse brain after intracerebroventricular CRH injections. As shown by immunohistochemistry and confocal microscopy analysis, CRH administration increased p-ERK1/2 levels specifically in the CA3 and CA1 hippocampal subfields and basolateral complex of the amygdala, both structures related to external environmental information processing and behavioral aspects of stress. Other regions such as hypothalamic nuclei and the central nucleus of the amygdala, also related to central CRH system but involved in the processing of the ascending visceral information and neuroendocrine-autonomic response to stress, did not show CRH-mediated ERK1/2 activation. To dissect the involvement of CRH receptor 1 (CRHR1) and CRHR2, we used conditional knockout mice in which Crhr1 is inactivated in the anterior forebrain and limbic structures. The conditional genetic ablation of Crhr1 inhibited the p-ERK1/2 increase, underlining the involvement of CRHR1 in the CRH-mediated activation. These findings underscore the fact that CRH activates p-ERK1/2 through CRHR1 only in selected brain regions, pointing to a specific role of this pathway in mediating behavioral adaptation to stress.


Asunto(s)
Encéfalo/enzimología , Hormona Liberadora de Corticotropina/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Animales , Activación Enzimática , Hipotálamo/enzimología , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Especificidad de Órganos , Prosencéfalo/enzimología
12.
Eur J Neurosci ; 20(2): 566-74, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15233766

RESUMEN

Prominent local decreases in blood oxygenation level (BOLD) can be observed by functional magnetic resonance imaging (fMRI) upon acoustic stimulation during sleep. The goal of this study was to further characterize this BOLD signal decrease with respect to corresponding neurophysiological phenomena using a simultaneous electroencephalography (EEG)/fMRI approach in sleeping human subjects. Healthy volunteers were subjected to acoustic stimulation during non-rapid eye movement (NREM) sleep. On the basis of statistical parametric maps, the correlations between the fMRI response (both amplitude and extent of the BOLD response) and the concomittant changes in the EEG (delta power and K-complexes) were calculated. Amplitude and extent of the stimulus-induced negative BOLD effect correlated positively with measures of EEG synchronization, namely an increase in the number of K-complexes and EEG delta power. Stimulus-induced BOLD decreases were most prominent during light (stage 2) NREM sleep and disappeared during slow wave sleep, indicating an influence of the baseline degree of hyperpolarization. Our observations provide first evidence that 'negative' BOLD signal changes during human sleep are associated with electrophysiological indicators of altered neuronal activity. Increased number of K-complexes and delta power reflecting hyperpolarization suggests true cortical deactivation upon stimulus presentation. This sleep stage-dependent deactivation might serve to protect the brain from arousing stimuli, particularly during the light phases of sleep shortly after sleep onset.


Asunto(s)
Circulación Cerebrovascular/fisiología , Imagen por Resonancia Magnética/métodos , Oxígeno/sangre , Transducción de Señal/fisiología , Sueño/fisiología , Estimulación Acústica/métodos , Adulto , Mapeo Encefálico , Electroencefalografía/métodos , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Masculino , Polisomnografía/métodos , Vigilia/fisiología
13.
Alcohol Clin Exp Res ; 28(3): 424-32, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15084900

RESUMEN

BACKGROUND: The high rate of comorbidity between alcoholism and anxiety disorders suggests some causal link. This study used the startle reflex to investigate whether increased reactivity to stimuli inducing fear or related affective states might be one mechanism by which a family genetic risk promotes the development of alcohol use disorders. METHODS: Thirty-one sons of alcoholics (PH+) were recruited from the participants of a longitudinal epidemiologic survey representative of the Munich area population between 18 and 25 years. Thirty male low-risk participants without parental alcoholism (PH-) were matched for age and history of psychiatric disorders. The baseline acoustic startle reflex was elicited before and after subjects drank 0.6 g/kg ethanol or placebo in a randomized, double-blind, placebo-controlled crossover design. Thereafter, the startle response was investigated while the subjects' affective state was manipulated by announcement of aversive electric finger stimuli to induce fear potentiation and by presentation of photographic slides previously rated to be pleasant, unpleasant, or neutral in their emotional valence. RESULTS: Plain startle response was lower in PH+ than PH- participants and was equally dampened by alcohol in PH+ and PH- subjects. Threat of finger shocks increased the startle response to the same extent in both groups. This fear potentiation effect was significantly attenuated by alcohol given on the second experimental day but not if alcohol was administered first and placebo on the second day. Pleasant and unpleasant slides decreased and increased startle response, respectively, and this effect was influenced by neither risk group nor alcohol. CONCLUSIONS: The acoustic startle reflex seems to be reduced in sons of alcoholics. The nonsignificant results during startle modification do not support the concept of increased reactivity to anxiety-related environmental stimuli as a mechanism promoting alcohol use disorders in subjects at increased family genetic risk for alcoholism.


Asunto(s)
Alcoholismo/genética , Ansiedad/genética , Etanol/farmacología , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/genética , Estimulación Acústica/métodos , Adolescente , Adulto , Alcoholismo/complicaciones , Alcoholismo/psicología , Análisis de Varianza , Ansiedad/inducido químicamente , Ansiedad/complicaciones , Estudios Cruzados , Método Doble Ciego , Padre/psicología , Humanos , Masculino , Núcleo Familiar/psicología
14.
Neuropsychopharmacology ; 28(2): 235-43, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12589376

RESUMEN

The neuroendocrine and behavioral effects of chronic paroxetine treatment were investigated in two rat lines selectively bred for high anxiety-related behavior (HAB) or low anxiety-related behavior (LAB) emotionality. In addition to a characteristic behavioral phenotype with markedly passive stress-coping strategies, HAB rats show a hypothalamic vasopressinergic hyperdrive that is causally related to hypothalamic-pituitary-adrenocortical dysregulation as demonstrated in the combined dexamethasone (DEX)/corticotropin-releasing hormone (CRH) test. A total of 8 weeks of chronic paroxetine treatment induced a more active coping strategy in the forced swim test in HAB rats only. In contrast, paroxetine-treated LAB rats did not change their swimming behavior. To investigate the neuroendocrine alterations linked to these behavioral changes, a combined DEX/CRH test was performed. In HAB rats, the paroxetine-induced behavioral changes towards more active coping strategies were accompanied by a normalization of the CRH-stimulated increase in corticotropin (ACTH) and corticosterone secretion. Concomitantly, the hypothalamic vasopressinergic hyperdrive was found to be reduced in HAB but not LAB rats, as indicated by a decrease in vasopressin mRNA expression, whereas vasopressin 1a receptor binding was unaffected. These findings provide the first evidence that the vasopressinergic system is likely to be critically involved in the behavioral and neuroendocrine effects of antidepressant drugs. This novel mechanism of action of paroxetine on vasopressin gene regulation renders vasopressinergic neuronal circuits a promising target for the development of more causal antidepressant treatment strategies.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Modelos Animales de Enfermedad , Hipotálamo/efectos de los fármacos , Sistemas Neurosecretores/efectos de los fármacos , Paroxetina/administración & dosificación , Vasopresinas/biosíntesis , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Hipotálamo/metabolismo , Masculino , Sistemas Neurosecretores/metabolismo , ARN Mensajero/biosíntesis , Ratas , Especificidad de la Especie , Vasopresinas/antagonistas & inhibidores
15.
Neuroimage ; 16(1): 251-8, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11969332

RESUMEN

Although there is evidence that acoustic stimuli are processed differently during sleep and wakefulness, little is known about the underlying neuronal mechanisms. In the present study, the processing of an acoustic stimulus was investigated during different non rapid eye movement (NREM) sleep stages using a combined EEG/fMRI approach in healthy human volunteers: A text stimulus was presented to sleep-deprived subjects prior to and after the onset of sleep, and single-slice silent fMRI were acquired. We found significantly different blood oxygenation level-dependent (BOLD) contrast responses during sleep compared to wakefulness. During NREM sleep stages 1 and 2 and during slow wave sleep (SWS) we observed reduced activation in the auditory cortex and a pronounced negative signal in the visual cortex and precuneus. Acoustic stimulation during sleep was accompanied by an increase in EEG frequency components in the low delta frequency range. Provided that neurovascular coupling is not altered during sleep, the negative transmodal BOLD response which is most pronounced during NREM sleep stages 1 and 2 reflects a deactivation predominantly in the visual cortex suggesting that this decrease in neuronal activity protects the brain from the arousing effects of external stimulation during sleep not only in the primary targeted sensory cortex but also in other brain regions.


Asunto(s)
Percepción Auditiva/fisiología , Encéfalo/fisiología , Sueño/fisiología , Percepción Visual/fisiología , Estimulación Acústica , Adolescente , Adulto , Corteza Auditiva/irrigación sanguínea , Corteza Auditiva/fisiología , Circulación Cerebrovascular/fisiología , Interpretación Estadística de Datos , Electrocardiografía , Electroencefalografía , Electrofisiología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Oxígeno/sangre , Polisomnografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA