Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Psychiatry ; 23(1): 331, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37170109

RESUMEN

BACKGROUND: Major depressive disorder (MDD) is a heterogeneous psychiatric disorder. Childhood trauma (CT, emotional/physical/sexual abuse or neglect before the age of 18) is one of the largest and most consistent risk factors for development and poor course of MDD. Overactivity of the HPA-axis and the stress hormone cortisol is thought to play a role in the vulnerability for MDD following exposure to CT. Rodent experiments showed that antagonism of the glucocorticoid receptor (GR) at adult age reversed the effects of early life stress. Similarly, we aim to target MDD in individuals with CT exposure using the GR antagonist mifepristone. METHODS: The RESET-medication study is a placebo-controlled double-blind randomized controlled trial (RCT) which aims to include 158 adults with MDD and CT. Participants will be randomized (1:1) to a 7-day treatment arm of mifepristone (1200 mg/day) or a control arm (placebo). Participants are allowed to receive usual care for MDD including antidepressants. Measurements include three face-to-face meetings at baseline (T0), day 8 (T1), week 6 (T2), and two online follow-up meetings at 12 weeks (T3) and 6 months (T4). A subgroup of participants (N = 80) are included in a fMRI sub-study (T0, T2). The main study outcome will be depressive symptom severity as measured with the Inventory of Depressive Symptomatology-Self Rated (IDS-SR) at T2. Secondary outcomes include, among others, depressive symptom severity at other time points, disability, anxiety, sleep and subjective stress. To address underlying mechanisms mifepristone plasma levels, cortisol, inflammation, epigenetic regulation and fMRI measurements are obtained. DISCUSSION: The RESET-medication study will provide clinical evidence whether GR antagonism is a disease-modifying treatment for MDD in individuals exposed to CT. If effective, this hypothesis-driven approach may extend to other psychiatric disorders where CT plays an important role. TRIAL REGISTRATION: The trial protocol has been registered 01-02-2022 on ClinicalTrials.gov with ID "NCT05217758".


Asunto(s)
Experiencias Adversas de la Infancia , Trastorno Depresivo Mayor , Mifepristona , Humanos , Experiencias Adversas de la Infancia/psicología , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/etiología , Trastorno Depresivo Mayor/psicología , Hidrocortisona , Mifepristona/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores de Glucocorticoides/antagonistas & inhibidores , Resultado del Tratamiento , Adulto
2.
Front Neuroendocrinol ; 49: 124-145, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29428549

RESUMEN

Bruce McEwen's discovery of receptors for corticosterone in the rat hippocampus introduced higher brain circuits in the neuroendocrinology of stress. Subsequently, these receptors were identified as mineralocorticoid receptors (MRs) that are involved in appraisal processes, choice of coping style, encoding and retrieval. The MR-mediated actions on cognition are complemented by slower actions via glucocorticoid receptors (GRs) on contextualization, rationalization and memory storage of the experience. These sequential phases in cognitive performance depend on synaptic metaplasticity that is regulated by coordinate MR- and GR activation. The receptor activation includes recruitment of coregulators and transcription factors as determinants of context-dependent specificity in steroid action; they can be modulated by genetic variation and (early) experience. Interestingly, inflammatory responses to damage seem to be governed by a similarly balanced MR:GR-mediated action as the initiating, terminating and priming mechanisms involved in stress-adaptation. We conclude with five questions challenging the MR:GR balance hypothesis.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Plasticidad Neuronal/fisiología , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Estrés Psicológico/metabolismo , Animales , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Estrés Psicológico/complicaciones , Estrés Psicológico/fisiopatología
3.
Mol Psychiatry ; 21(12): 1733-1739, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26976039

RESUMEN

Maladaptive glucocorticoid effects contribute to stress-related psychopathology. The glucocorticoid receptor (GR) that mediates many of these effects uses multiple signaling pathways. We have tested the hypothesis that manipulation of downstream factors ('coregulators') can abrogate potentially maladaptive GR-mediated effects on fear-motivated behavior that are linked to corticotropin releasing hormone (CRH). For this purpose the expression ratio of two splice variants of steroid receptor coactivator-1 (SRC-1) was altered via antisense-mediated 'exon-skipping' in the central amygdala of the mouse brain. We observed that a change in splicing towards the repressive isoform SRC-1a strongly reduced glucocorticoid-induced responsiveness of Crh mRNA expression and increased methylation of the Crh promoter. The transcriptional GR target gene Fkbp5 remained responsive to glucocorticoids, indicating gene specificity of the effect. The shift of the SRC-1 splice variants altered glucocorticoid-dependent exploratory behavior and attenuated consolidation of contextual fear memory. In conclusion, our findings demonstrate that manipulation of GR signaling pathways related to the Crh gene can selectively diminish potentially maladaptive effects of glucocorticoids.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Coactivador 1 de Receptor Nuclear/metabolismo , Empalme Alternativo , Amígdala del Cerebelo , Animales , Corticosterona/metabolismo , Miedo , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/metabolismo , Ratones , Coactivador 1 de Receptor Nuclear/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de ARN , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Esteroides , Proteínas de Unión a Tacrolimus/metabolismo
4.
Mol Psychiatry ; 18(9): 993-1005, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22925833

RESUMEN

Glucocorticoids (GCs) secreted after stress reduce adult hippocampal neurogenesis, a process that has been implicated in cognitive aspects of psychopathology, amongst others. Yet, the exact role of the GC receptor (GR), a key mediator of GC action, in regulating adult neurogenesis is largely unknown. Here, we show that GR knockdown, selectively in newborn cells of the hippocampal neurogenic niche, accelerates their neuronal differentiation and migration. Strikingly, GR knockdown induced ectopic positioning of a subset of the new granule cells, altered their dendritic complexity and increased their number of mature dendritic spines and mossy fiber boutons. Consistent with the increase in synaptic contacts, cells with GR knockdown exhibit increased basal excitability parallel to impaired contextual freezing during fear conditioning. Together, our data demonstrate a key role for the GR in newborn hippocampal cells in mediating their synaptic connectivity and structural as well as functional integration into mature hippocampal circuits involved in fear memory consolidation.


Asunto(s)
Hipocampo/citología , Motivación/genética , Neurogénesis/genética , Neuronas/fisiología , Receptores de Glucocorticoides/deficiencia , Animales , Movimiento Celular/genética , Condicionamiento Clásico/fisiología , Corticosterona/metabolismo , Dendritas/metabolismo , Dendritas/ultraestructura , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Miedo , Vectores Genéticos/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Terminales Presinápticos/metabolismo , ARN Interferente Pequeño/metabolismo , Radioinmunoensayo
5.
Bone Rep ; 20: 101750, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38566930

RESUMEN

Age-associated osteoporosis (AAOP) poses a significant health burden, characterized by increased fracture risk due to declining bone mass and strength. Effective prevention and early treatment strategies are crucial to mitigate the disease burden and the associated healthcare costs. Current therapeutic approaches effectively target the individual contributing factors to AAOP. Nonetheless, the management of AAOP is complicated by the multitude of variables that affect its development. Main intrinsic and extrinsic factors contributing to AAOP risk are reviewed here, including mechanical unloading, nutrient deficiency, hormonal disbalance, disrupted metabolism, cognitive decline, inflammation and circadian disruption. Furthermore, it is discussed how these can be targeted for prevention and treatment. Although valuable as individual targets for intervention, the interconnectedness of these risk factors result in a unique etiology for every patient. Acknowledgement of the multifaceted nature of AAOP will enable the development of more effective and sustainable management strategies, based on a holistic, patient-centered approach.

6.
J Neuroendocrinol ; 36(2): e13367, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38281730

RESUMEN

The hypothalamic paraventricular nucleus (PVN) is a highly complex brain region that is crucial for homeostatic regulation through neuroendocrine signaling, outflow of the autonomic nervous system, and projections to other brain areas. In the past years, single-cell datasets of the hypothalamus have contributed immensely to the current understanding of the diverse hypothalamic cellular composition. While the PVN has been adequately classified functionally, its molecular classification is currently still insufficient. To address this, we created a detailed atlas of PVN transcriptomic cell types by integrating various PVN single-cell datasets into a recently published hypothalamus single-cell transcriptome atlas. Furthermore, we functionally profiled transcriptomic cell types, based on relevant literature, existing retrograde tracing data, and existing single-cell data of a PVN-projection target region. Finally, we validated our findings with immunofluorescent stainings. In our PVN atlas dataset, we identify the well-known different neuropeptide types, each composed of multiple novel subtypes. We identify Avp-Tac1, Avp-Th, Oxt-Foxp1, Crh-Nr3c1, and Trh-Nfib as the most important neuroendocrine subtypes based on markers described in literature. To characterize the preautonomic functional population, we integrated a single-cell retrograde tracing study of spinally projecting preautonomic neurons into our PVN atlas. We identify these (presympathetic) neurons to cocluster with the Adarb2+ clusters in our dataset. Further, we identify the expression of receptors for Crh, Oxt, Penk, Sst, and Trh in the dorsal motor nucleus of the vagus, a key region that the pre-parasympathetic PVN neurons project to. Finally, we identify Trh-Ucn3 and Brs3-Adarb2 as some centrally projecting populations. In conclusion, our study presents a detailed overview of the transcriptomic cell types of the murine PVN and provides a first attempt to resolve functionality for the identified populations.


Asunto(s)
Núcleo Hipotalámico Paraventricular , Transcriptoma , Ratones , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Análisis de Expresión Génica de una Sola Célula , Hipotálamo/metabolismo , Perfilación de la Expresión Génica
7.
Prog Retin Eye Res ; 87: 100994, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34280556

RESUMEN

The choroid is a key player in maintaining ocular homeostasis and plays a role in a variety of chorioretinal diseases, many of which are poorly understood. Recent advances in the field of single-cell RNA sequencing have yielded valuable insights into the properties of choroidal endothelial cells (CECs). Here, we review the role of the choroid in various physiological and pathophysiological mechanisms, focusing on the role of CECs. We also discuss new insights regarding the phenotypic properties of CECs, CEC subpopulations, and the value of measuring transcriptomics in primary CEC cultures derived from post-mortem eyes. In addition, we discuss key phenotypic, structural, and functional differences that distinguish CECs from other endothelial cells such as retinal vascular endothelial cells. Understanding the specific clinical and molecular properties of the choroid will shed new light on the pathogenesis of the broad clinical range of chorioretinal diseases such as age-related macular degeneration, central serous chorioretinopathy and other diseases within the pachychoroid spectrum, uveitis, and diabetic choroidopathy. Although our knowledge is still relatively limited with respect to the clinical features and molecular pathways that underlie these chorioretinal diseases, we summarise new approaches and discuss future directions for gaining new insights into these sight-threatening diseases and highlight new therapeutic strategies such as pluripotent stem cell‒based technologies and gene therapy.


Asunto(s)
Coriorretinopatía Serosa Central , Enfermedades de la Coroides , Degeneración Macular , Coroides/irrigación sanguínea , Enfermedades de la Coroides/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Angiografía con Fluoresceína , Humanos , Degeneración Macular/genética , Tomografía de Coherencia Óptica
8.
Psychoneuroendocrinology ; 132: 105334, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34225183

RESUMEN

INTRODUCTION: Cushing's disease (CD) is a rare and severe endocrine disease characterized by hypercortisolemia. Previous studies have found structural brain alterations in remitted CD patients compared to healthy controls, specifically in the anterior cingulate cortex (ACC). However, potential mechanisms through which these persistent alterations may have occurred are currently unknown. METHODS: Structural 3T MRI's from 25 remitted CD patients were linked with gene expression data from neurotypical donors, derived from the Allen Human Brain Atlas. Differences in gene expression between the ACC and an unaffected control cortical region were examined, followed by a Gene Ontology (GO) enrichment analysis. A cell type enrichment analysis was conducted on the differentially expressed genes, and a disease association enrichment analysis was conducted to determine possible associations between differentially expressed genes and specific diseases. Subsequently, cortisol sensitivity of these genes in existing datasets was examined. RESULTS: The gene expression analysis identified 300 differentially expressed genes in the ACC compared to the cortical control region. GO analyses found underexpressed genes to represent immune function. The cell type specificity analysis indicated that underexpressed genes were enriched for deactivated microglia and oligodendrocytes. Neither significant associations with diseases, nor evidence of cortisol sensitivity with the differentially expressed genes were found. DISCUSSION: Underexpressed genes in the ACC, the area vulnerable to permanent changes in remitted CD patients, were often associated with immune functioning. The specific lack of deactivated microglia and oligodendrocytes implicates protective effects of these cell types against the long-term effects of cortisol overexposure.


Asunto(s)
Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Corteza Cerebral/patología , Sustancia Gris/patología , Humanos , Hidrocortisona/metabolismo , Inmunidad/genética , Microglía/fisiología , Oligodendroglía/fisiología , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/patología , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/fisiopatología
9.
Transl Psychiatry ; 10(1): 293, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32826851

RESUMEN

Long-term remitted Cushing's disease (LTRCD) patients commonly continue to present persistent psychological and cognitive deficits, and alterations in brain function and structure. Although previous studies have conducted gray matter volume analyses, assessing cortical thickness and surface area of LTRCD patients may offer further insight into the neuroanatomical substrates of Cushing's disease. Structural 3T magnetic resonance images were obtained from 25 LTRCD patients, and 25 age-, gender-, and education-matched healthy controls (HCs). T1-weighted images were segmented using FreeSurfer software to extract mean cortical thickness and surface area values of 68 cortical gray matter regions and two whole hemispheres. Paired sample t tests explored differences between the anterior cingulate cortex (ACC; region of interest), and the whole brain. Validated scales assessed psychiatric symptomatology, self-reported cognitive functioning, and disease severity. After correction for multiple comparisons, ROI analyses indicated that LTRCD-patients showed reduced cortical thickness of the left caudal ACC and the right rostral ACC compared to HCs. Whole-brain analyses indicated thinner cortices of the left caudal ACC, left cuneus, left posterior cingulate cortex, right rostral ACC, and bilateral precuneus compared to HCs. No cortical surface area differences were identified. Cortical thickness of the left caudal ACC and left cuneus were inversely associated with anxiety symptoms, depressive symptoms, and disease duration, although certain associations did not persist after correction for multiple testing. In six of 68 regions examined, LTRCD patients had reduced cortical thickness in comparison to HCs. Cortical thickness of the left caudal ACC was inversely associated with disease duration. This suggests that prolonged and excessive exposure to glucocorticoids may be related to cortical thinning of brain structures involved in emotional and cognitive processing.


Asunto(s)
Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Encéfalo , Corteza Cerebral/diagnóstico por imagen , Sustancia Gris/diagnóstico por imagen , Giro del Cíngulo , Humanos , Imagen por Resonancia Magnética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/diagnóstico por imagen
10.
Endocrinology ; 149(2): 725-32, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18006628

RESUMEN

Nuclear receptor coregulators are proteins that modulate the transcriptional activity of steroid receptors and may explain cell-specific effects of glucocorticoid receptor action. Based on the uneven distribution of a number of coregulators in CRH-expressing cells in the hypothalamus of the rat brain, we tested the hypothesis that these proteins are involved as mediators in the glucocorticoid-induced repression of the CRH promoter. Therefore, we assessed the role of coregulator proteins on both induction and repression of CRH in the AtT-20 cell line, a model system for CRH repression by glucocorticoids. The steroid receptor coactivator 1a (SRC1a), SRC-1e, nuclear corepressor (N-CoR), and silencing mediator of the retinoid and thyroid hormone receptor (SMRT) were studied in this system. We show that the concentration of glucocorticoid receptor and the type of ligand, i.e. corticosterone or dexamethasone, determines the repression. Furthermore, overexpression of SRC1a, but not SRC1e, increased both efficacy and potency of the glucocorticoid receptor-mediated repression of the forskolin-induced CRH promoter. Unexpectedly, cotransfection of the corepressors N-CoR and SMRT did not affect the corticosterone-dependent repression but resulted in a marked decrease of the forskolin stimulation of the CRH gene. Altogether, our data demonstrate that 1) the concentration of the receptor, 2) the type of ligand, and 3) the coregulator recruited all determine the expression and the repression of the CRH gene. We conclude that modulation of coregulator activity may play a role in the control of the hypothalamus-pituitary-adrenal axis.


Asunto(s)
Hormona Liberadora de Corticotropina/genética , Histona Acetiltransferasas/metabolismo , Hipotálamo/fisiología , Proteínas Nucleares/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Colforsina/farmacología , Corticosterona/metabolismo , Corticosterona/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dexametasona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glucocorticoides/farmacología , Histona Acetiltransferasas/genética , Ratones , Proteínas Nucleares/genética , Co-Represor 1 de Receptor Nuclear , Co-Represor 2 de Receptor Nuclear , Coactivador 1 de Receptor Nuclear , Regiones Promotoras Genéticas/fisiología , Proteínas Represoras/genética , Factores de Transcripción/genética , Transfección
11.
Neuroscience ; 138(3): 891-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16310313

RESUMEN

Glucocorticoid hormones modulate brain function and as such are crucial for responding and adjusting to physical and psychological stressors. Their effects are mediated via mineralo- and glucocorticoid receptors, which in large measure act as transcription factors to modulate transcription of target genes, in a receptor-, cell-, and state-specific manner. The nature and magnitude of these transcriptional effects depend on the presence and activity of downstream proteins, such as steroid receptor coactivators and corepressors (together: coregulators), many of which are expressed in the brain. We address the role of coregulators for mineralo- and glucocorticoid receptor-mediated modulation of gene transcription. We first address evidence from cell lines for the importance of coregulator stoichiometry for steroid signaling. The in vivo importance of coregulators-when possible specifically for glucocorticoid signaling in the brain-is discussed based on knockout mice, transient knockdown of steroid receptor coactivators, and distribution and regulation of coactivator expression in the brain. We conclude that for a better understanding of modulation of brain function by glucocorticoids, it is necessary to take into account the role of coregulators, and to assess their importance relative to changes in hormone levels and receptor expression.


Asunto(s)
Encéfalo/fisiología , Receptores de Glucocorticoides/fisiología , Receptores de Mineralocorticoides/fisiología , Estrés Fisiológico/fisiopatología , Animales , Encéfalo/fisiopatología , Regulación de la Expresión Génica , Homeostasis , Cinética , Transducción de Señal , Esteroides/fisiología , Transcripción Genética
12.
J Neurosci ; 20(9): 3129-38, 2000 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10777776

RESUMEN

Glucocorticoids (GCs) are critical to learning and memory, in large part because of their actions in the hippocampus. Chronic high levels of GCs have profound effects on hippocampal structure and function and can even result in irreversible neurodegeneration. Hippocampal GC actions are mediated by intracellular receptors that modulate the transcription of specific target genes. In a screen for genes repressed by GCs in rat hippocampus, we identified plasma membrane calcium pump isoform 1 (PMCA1), a plasma membrane calcium ATPase. In Northern blots, PMCA1 was repressed approximately 33% after a high, but not a low dose of the GC, corticosterone (B), suggesting glucocorticoid (but not mineralocorticoid) receptor-mediated repression. Furthermore, in situ hybridization demonstrated that B significantly downregulated PMCA1 mRNA in all brain regions examined. Repression of PMCA1 was also observed in cultured hippocampal neurons, but only when the cells were in the differentiated state. Stress also repressed PMCA1 expression in hippocampus of adrenal-intact animals, and a clear inverse correlation between B level and PMCA1 mRNA could be discerned. However, other non-B-dependent factors appeared to be involved in the response of PMCA1 to stress because, unlike exogenous B, cold stress did not repress PMCA1 in brain regions other than hippocampus. Moreover, in the presence of constant B (B-replaced, adrenalectomized animals), cold stress led to increased hippocampal PMCA1 expression. These observations suggest that repression of PMCA1 represents one molecular mechanism by which corticosteroids regulate Ca(2+) homeostasis and hence influence neuronal activity. Moreover, other stress-related neurohumoral factors appear to counter the repressive effects of B. Defects in the balance between GC-mediated and non-GC-mediated effects on PMCA1 expression may have adverse effects on neuronal function and ultimately result in irreversible neuronal damage.


Asunto(s)
Antiinflamatorios/farmacología , ATPasas Transportadoras de Calcio/efectos de los fármacos , Corticosterona/farmacología , Expresión Génica/efectos de los fármacos , Estrés Fisiológico/metabolismo , Adrenalectomía , Animales , ATPasas Transportadoras de Calcio/genética , ATPasas Transportadoras de Calcio/metabolismo , Proteínas de Transporte de Catión , Células Cultivadas , Expresión Génica/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , ATPasas Transportadoras de Calcio de la Membrana Plasmática , Ratas , Ratas Sprague-Dawley
13.
Endocrinology ; 146(3): 1372-81, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15564338

RESUMEN

Glucocorticoid hormones are released after activation of the hypothalamus-pituitary-adrenal (HPA) axis and in the brain can modulate synaptic plasticity and memory formation. Clear individual differences in spatial learning and memory in the water maze allowed classification of groups of young (3 months) and aged (24 months) male Wistar rats as superior and inferior learners. We tested 1) whether measures of HPA activity are associated with cognitive functions and aging and 2) whether correlations of these measures depend on age and learning performance. Basal ACTH, but not corticosterone, was increased in aged rats, with the stress-induced ACTH response exaggerated in aged-inferior learners. Aged-superior learners had lower expression of glucocorticoid receptor and CRH mRNA in the parvocellular paraventricular nucleus of the hypothalamus compared with all other groups. Hippocampal mineralocorticoid receptor and glucocorticoid receptor mRNAs differed modestly between groups, but steroid receptor coactivator and heat-shock-protein 90 mRNAs were not different. Strikingly, correlations between HPA axis markers were dependent on grouping animals according to learning performance or age. CRH mRNA correlated with ACTH only in aged animals. Parvocellular arginine vasopressin mRNA was negatively correlated to basal corticosterone, except in aged-inferior learners. Corticosteroid receptor mRNA expression showed a number of correlations with other HPA axis regulators specifically in superior learners. In summary, the relationships between HPA axis markers differ for subgroups of animals. These distinct interdependencies may reflect adjusted set-points of the HPA axis, resulting in adaptation (or maladaptation) to the environment and, possibly, an age-independent determination of learning ability.


Asunto(s)
Glándulas Suprarrenales/fisiología , Envejecimiento , Hipotálamo/fisiología , Aprendizaje , Hipófisis/fisiología , Sistema Hipófiso-Suprarrenal , Animales , Arginina Vasopresina/metabolismo , Corticosterona/metabolismo , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Hibridación in Situ , Masculino , Aprendizaje por Laberinto , Plásmidos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Factores de Tiempo
14.
Endocrinology ; 146(12): 5587-95, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16150912

RESUMEN

The synthetic glucocorticoid dexamethasone (dex) blocks stress-induced hypothalamic-pituitary-adrenal (HPA) activation primarily at the level of the anterior pituitary because multidrug resistance P-glycoprotein hampers its penetration in the brain. Here, we tested the hypothesis that central components of the HPA axis would escape dex suppression under conditions of potent peripheral glucocorticoid action. We subchronically treated rats with low or high doses of dex. The animals were subjected on the last day of treatment for 30 min to a restraint stressor after which central and peripheral markers of HPA axis activity were measured. Basal and stress-induced corticosterone secretion, body weight gain, adrenal and thymus weight, as well as proopiomelanocortin mRNA in the anterior pituitary were reduced in a dose-dependent manner by dex administered either 5 d sc or 3 wk orally. In the brain, the highest dose dex suppressed CRH mRNA and CRH heteronuclear RNA in the paraventricular nucleus (PVN). However, in the peripherally active low-dose range of dex CRH mRNA and heteronuclear RNA showed resistance to suppression, and CRH mRNA expression in the PVN was in fact enhanced under the long-term treatment condition. In the PVN, c-fos mRNA was suppressed by the highest dose of dex, but this effect showed a degree of resistance after long-term oral treatment. c-fos mRNA responses in the anterior pituitary followed those in PVN and reflect central drive of the HPA axis even if corticosterone responses are strongly reduced. The results support the concept that low doses of dex can create a hypocorticoid state in the brain.


Asunto(s)
Corticoesteroides/deficiencia , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dexametasona/administración & dosificación , Administración Oral , Adrenalectomía , Animales , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/genética , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inyecciones Subcutáneas , Masculino , Núcleo Hipotalámico Paraventricular/metabolismo , Adenohipófisis/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Nuclear Heterogéneo/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Restricción Física , Estrés Fisiológico/etiología , Estrés Fisiológico/metabolismo
15.
Endocrinology ; 146(3): 1438-48, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15564339

RESUMEN

The mechanisms of receptor- and cell-specific effects of the adrenal corticosteroid hormones via mineralo- (MRs) and glucocorticoid receptors (GRs) are still poorly understood. Because the expression levels of two splice variants of the steroid receptor coactivator-1 (SRC-1) 1a and 1e, can differ significantly in certain cell populations, we tested the hypothesis that their relative abundance could determine cell- and receptor-specific effects of corticosteroid receptor-mediated transcription. In transient transfections, we demonstrate three novel types of SRC-1a- and SRC-1e-specific effects for corticosteroid receptors. One is promoter dependence: SRC-1e much more potently coactivated transcription from several multiple response element-containing promoters. Mammalian 1-hydrid studies indicated that this likely does not involve promoter-specific coactivator recruitment. Endogenous phenylethanolamine-N-methyltransferase mRNA induction via GRs was also differentially affected by the splice variants. Another type is receptor specificity: responses mediated by the N-terminal part of the MR, but not the GR, were augmented by SRC-1e at synergizing response elements. SRC fragment SRC(988-1240) by the MR but not the GR N-terminal fragment in a 1-hybrid assay. The last type, for GRs, is ligand dependence. Due to effects on partial agonism of RU486-activated GRs, different ratios of SRC-1a and 1e can lead to large differences in the extent of antagonism of RU486 on GR-mediated transcription. Furthermore, we show that SRC-1e but not SRC-1a mRNA expression was regulated in the pituitary by corticosterone. We conclude that the cellular differences in SRC-1a to SRC-1e ratio demonstrated in vivo might be involved in cell-specific responses to corticosteroids in a promoter- and ligand-dependent way.


Asunto(s)
Receptores de Esteroides/metabolismo , Transducción de Señal , Factores de Transcripción/biosíntesis , Factores de Transcripción/química , Empalme Alternativo , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Corticosterona/metabolismo , Genes Reporteros , Histona Acetiltransferasas , Humanos , Hibridación in Situ , Ligandos , Modelos Biológicos , Coactivador 1 de Receptor Nuclear , Plásmidos/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factores de Transcripción/genética , Transcripción Genética , Transfección
16.
Brain Res ; 1059(2): 113-21, 2005 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-16212947

RESUMEN

The two structurally related nuclear receptor corepressor (N-CoR) and silencing mediator of retinoid and thyroid receptors (SMRT) proteins have been found to differentially affect the transcriptional activity of numerous nuclear receptors, such as thyroid hormone, retinoic acid and steroid receptors. Because of the numerous effects mediated by nuclear receptors in brain, it is of interest to extend these in vitro data and to explore the cellular distribution of both corepressors in brain tissue. We therefore examined, using in situ hybridisation, whether the relative abundance of these two functionally distinct corepressors differed in rat brain and pituitary. We find that although both N-CoR and SMRT transcripts are ubiquitously expressed in brain, striking differences in their respective levels of expression could be observed in discrete areas of brain stem, thalamus, hypothalamus and hippocampus. Using dual-label immunofluorescence, we examined in selected glucocorticoid sensitive areas involved in the regulation of the hypothalamus-pituitary-adrenal axis activity, the respective protein abundance of N-CoR and SMRT. Protein abundance was largely concurrent with the mRNA expression levels, with SMRT relatively more abundant in hypothalamus and brain stem areas. Colocalisation of N-CoR and SMRT was demonstrated by confocal microscopy in most areas studied. Taken together, these findings are consistent with the idea that the uneven neuroanatomical distribution of N-CoR and SMRT protein may contribute to the site-specific effects exerted by hormones, such as glucocorticoids, in the brain.


Asunto(s)
Mapeo Encefálico , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Hipófisis/metabolismo , Proteínas Represoras/metabolismo , Animales , Proteínas de Unión al ADN/genética , Sistema Hipotálamo-Hipofisario/metabolismo , Hibridación in Situ , Masculino , Proteínas Nucleares/genética , Co-Represor 1 de Receptor Nuclear , Co-Represor 2 de Receptor Nuclear , Sistema Hipófiso-Suprarrenal/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Proteínas Represoras/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Distribución Tisular , Transcripción Genética/genética , Transcripción Genética/fisiología
17.
Trends Endocrinol Metab ; 11(6): 245-8, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11273562

RESUMEN

Novel aspects of nuclear receptors and their function in brain were discussed at a recent Symposium in Oegstgeest, The Netherlands. Presentations covered the diversity of these receptors, their target genes, proteins involved in transcriptional regulation, functional consequences of nuclear receptor activation and their relevance for human pathology. By elucidating the signalling pathway of nuclear receptors in brain, potential targets for therapeutic treatment of brain disorders can be identified.


Asunto(s)
Encéfalo/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Humanos
18.
Endocrinology ; 141(6): 2192-9, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10830308

RESUMEN

Members of the p160 family of steroid receptor coactivator proteins mediate the stimulatory effects on gene transcription brought about by nuclear receptors, which comprise all steroid receptors. Using in situ hybridization we have examined the neuroanatomical distribution of the messenger RNAs (mRNAs) for two functionally distinct splice variants of Steroid Receptor Coactivator 1 (SRC-1/NCoA-1) and of Steroid Receptor Coactivator 2 (SRC-2/NCoA-2/GRIP-1/TIF-2). Transcripts encoding these coactivators show highly differential expression patterns. SRC-2 mRNA is expressed at very low levels in brain, but shows expression in the anterior pituitary. SRC-la and le mRNA are expressed in many brain areas, including hippocampus, amygdala, hypothalamus, basal ganglia, and isocortex. Striking differences between SRC-1a and le expression were observed in several brain nuclei. Relative levels of SRC-1a mRNA were much higher in anterior pituitary, and the arcuate, paraventricular and ventromedial nucleus of the hypothalamus, the locus coeruleus and the trigeminal motor nucleus, all important targets of steroid hormones in the brain. SRC-le mRNA showed modest elevation of relative expression in the caudal nucleus accumbens (shell), basolateral amygdala, and some thalamic nuclei. The differential and uneven neuroanatomical distribution of these coactivators may underlie diversity and cell-specificity of steroid receptor mediated signals in the brain.


Asunto(s)
Química Encefálica , Expresión Génica , Hipófisis/química , Factores de Transcripción/genética , Amígdala del Cerebelo/química , Animales , Ganglios Basales/química , Secuencia de Bases , Encéfalo/metabolismo , Corteza Cerebral/química , Hipocampo/química , Histona Acetiltransferasas , Hipotálamo/química , Hibridación in Situ , Masculino , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Sondas de Oligonucleótidos , Hipófisis/metabolismo , ARN Mensajero/análisis , ARN Mensajero/química , Ratas , Ratas Wistar , Distribución Tisular
19.
Endocrinology ; 142(6): 2686-94, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11356720

RESUMEN

In the present study, we investigated the role of the multidrug resistance (mdr) P-glycoprotein (Pgp) at the blood-brain barrier in the control of access of cortisol and corticosterone to the mouse and human brain. [(3)H]Cortisol poorly penetrated the brain of adrenalectomized wild-type mice, but the uptake was 3.5-fold enhanced after disruption of Pgp expression in mdr 1a(-/-) mice. In sharp contrast, treatment with [(3)H]corticosterone revealed high labeling of brain tissue without difference between both genotypes. Interestingly, human MDR1 Pgp also differentially transported cortisol and corticosterone. LLC-PK1 monolayers stably transfected with MDR1 complementary DNA showed polar transport of [(3)H]cortisol that could be blocked by a specific Pgp blocker, whereas [(3)H]corticosterone transport did not differ between transfected and host cells. Determination of the concentration of both steroids in extracts of human postmortem brain tissue using liquid chromatography mass spectrometry revealed that the ratio of corticosterone over cortisol in the brain was significantly increased relative to plasma. In conclusion, the data demonstrate that in both mouse and human brain the penetration of cortisol is less than that of corticosterone. This finding suggests a more prominent role for corticosterone in control of human brain function than hitherto recognized.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Encéfalo/metabolismo , Corticosterona/metabolismo , Hidrocortisona/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Amígdala del Cerebelo/metabolismo , Animales , Transporte Biológico , Línea Celular , Cerebelo/metabolismo , Cromatografía Liquida , Corticosterona/sangre , Resistencia a Múltiples Medicamentos , Epitelio/metabolismo , Expresión Génica , Hipocampo/metabolismo , Humanos , Hidrocortisona/sangre , Riñón , Masculino , Espectrometría de Masas , Ratones , Porcinos , Transfección , Tritio
20.
Endocrinology ; 139(4): 1789-93, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9528963

RESUMEN

Mice with a genetic disruption of the multiple drug resistance (mdr1a) gene were used to examine the effect of the absence of its drug-transporting P-glycoprotein product from the blood-brain barrier on the distribution and cell nuclear uptake of [3H]-dexamethasone in the brain. [3H]-dexamethasone (4 microg/kg mouse) was administered s.c. to adrenalectomized mdr1a (-/-) and mdr1a (+/+) mice. One hour later, the mice were decapitated, and the radioactivity was measured in homogenates of cerebellum, blood, and liver following extraction of the radioactive steroid. The frontal brain was cut in sections for autoradiography. In the cerebellum of the mdr1a mutants, the amount of [3H]-dexamethasone relative to blood was about 5-fold higher than observed in the controls, whereas the ratio in blood vs. liver was not different. Using autoradiography, it was found that brain areas expressing the glucocorticoid receptor (GR) in high abundance, such as the hippocampal cell fields and the paraventricular nucleus (PVN), showed a 10-fold increase in cell nuclear uptake of radiolabeled steroid. The amount of retained steroid increased toward levels observed in the pituitary, which contains a similar density of GRs. The [3H]-dexamethasone concentration in pituitary was not affected by mdr1a gene disruption. The GR messenger RNA expression pattern in hippocampus was not different between the wild types and mdr1a mutants, which rules out altered receptor expression as a cause of the enhanced dexamethasone uptake. In conclusion, the present study demonstrates that the brain is resistant to penetration by dexamethasone because of mdr1a activity at the level of the blood-brain barrier. The data support the concept of a pituitary site of action of dexamethasone in blockade of stress-induced ACTH release. Dexamethasone poorly substitutes for depletion of the endogenous glucocorticoid from the brain and therefore, in this tissue, may cause a condition resembling that of adrenalectomy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Barrera Hematoencefálica , Encéfalo/metabolismo , Dexametasona/farmacocinética , Glucocorticoides/farmacocinética , Receptores de Glucocorticoides/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adrenalectomía , Animales , Encéfalo/ultraestructura , Núcleo Celular/metabolismo , Cerebelo/metabolismo , Corticosterona/metabolismo , Dexametasona/sangre , Dexametasona/metabolismo , Expresión Génica , Hipocampo/metabolismo , Hibridación in Situ , Hígado/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Tritio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA