Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Front Neuroendocrinol ; 69: 101065, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37001566

RESUMEN

The two-hit stress model predicts that exposure to stress at two different time-points in life may increase or decrease the risk of developing stress-related disorders later in life. Most studies based on the two-hit stress model have investigated early postnatal stress as the first hit with adult stress as the second hit. Adolescence, however, represents another highly sensitive developmental window during which exposure to stressful events may affect programming outcomes following exposure to stress in adulthood. Here, we discuss the programming effects of different types of stressors (social and nonsocial) occurring during adolescence (first hit) and how such stressors affect the responsiveness toward an additional stressor occurring during adulthood (second hit) in rodents. We then provide a comprehensive overview of the potential mechanisms underlying interindividual and sex differences in the resilience/susceptibility to developing stress-related disorders later in life when stress is experienced in two different life stages.


Asunto(s)
Estrés Psicológico , Animales , Femenino , Masculino , Estrés Psicológico/complicaciones , Roedores , Factores Sexuales
2.
Psychosom Med ; 85(6): 551-560, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37234022

RESUMEN

OBJECTIVE: The objective of this study was to investigate whether placebo effect induced by pharmacological conditioning with intranasal insulin can affect glucose, insulin, C-peptide, hunger, and memory in patients with diabetes type 2 and healthy controls. METHODS: Placebo effect was induced by pharmacological conditioning. Thirty-two older patients (mean age = 68.3 years) with diabetes type 2 and age- and sex-matched thirty-two healthy older adults (mean age = 67.8 years) were randomly assigned to a conditioned or a control group. On day 1, conditioned group received six administrations of intranasal insulin with a conditioned stimulus (CS; smell of rosewood oil), whereas the control group received a placebo with the CS. On day 2, both groups received a placebo spray with the CS. Glucose, insulin, and C-peptide were repeatedly measured in blood. Hunger and memory were assessed with validated measures. RESULTS: Intranasal insulin stabilized dropping glucose levels in patients ( B = 0.03, SE = 0.02, p = .027) and healthy men ( B = 0.046, SE = 0.02, p = .021), and decreased C-peptide levels in healthy controls ( B = 0.01, SE = 0.001, p = .008). Conditioning also prevented the drop of glucose levels but only in men (both healthy and patients; B = 0.001, SE = 0.0003, p = .024). Conditioning significantly decreased hunger in healthy participants ( B = 0.31, SE = 0.09, p < .001). No effects were found on other measures. CONCLUSIONS: Placebo effect induced by conditioning with intranasal insulin modifies blood glucose levels and decreases hunger in older adults, but its effects depend on health status and sex. Insulin conditioning might be beneficial for groups suffering from intensive hunger but seems not be particularly suitable for blood glucose reduction. TRIAL REGISTRATION: Netherlands Trial Register, NL7783 ( https://www.trialregister.nl/trial/7783 ).


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulina , Masculino , Humanos , Anciano , Glucemia , Efecto Placebo , Péptido C/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucosa/farmacología , Glucosa/uso terapéutico , Estado de Salud , Método Doble Ciego , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico
3.
FASEB J ; 36(4): e22251, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35262955

RESUMEN

Glucocorticoids exert their pleiotropic effects by activating the glucocorticoid receptor (GR), which is expressed throughout the body. GR-mediated transcription is regulated by a multitude of tissue- and cell type-specific mechanisms, including interactions with other transcription factors such as the androgen receptor (AR). We previously showed that the transcription of canonical glucocorticoid-responsive genes is dependent on active androgen signaling, but the extent of this glucocorticoid-androgen crosstalk warrants further investigation. In this study, we investigated the overall glucocorticoid-androgen crosstalk in the hepatic transcriptome. Male mice were exposed to GR agonist corticosterone and AR antagonist enzalutamide in order to determine the extent of androgen-dependency after acute and chronic exposure. We found that a substantial proportion of the hepatic transcriptome is androgen-dependent after chronic exposure, while after acute exposure the transcriptomic effects of glucocorticoids are largely androgen-independent. We propose that prolonged glucocorticoid exposure triggers a gradual upregulation of AR expression, instating a situation of androgen dependence which is likely not driven by direct AR-GR interactions. This indirect mode of glucocorticoid-androgen interaction is in accordance with the absence of enriched AR DNA-binding near AR-dependent corticosterone-regulated genes after chronic exposure. In conclusion, we demonstrate that glucocorticoid effects and their interaction with androgen signaling are dependent on the duration of exposure and believe that our findings contribute to a better understanding of hepatic glucocorticoid biology in health and disease.


Asunto(s)
Andrógenos , Glucocorticoides , Andrógenos/metabolismo , Andrógenos/farmacología , Animales , Corticosterona/farmacología , Regulación de la Expresión Génica , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Masculino , Ratones , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
4.
Stress ; 26(1): 2275210, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37874158

RESUMEN

Glucocorticoid hormones are essential for health, but overexposure may lead to many detrimental effects, including metabolic, psychiatric, and bone disease. These effects may not only be due to increased overall exposure to glucocorticoids, but also to elevated hormone levels at the time of the physiological circadian trough of glucocorticoid levels. The late Mary Dallman developed a model that allows the differentiation between the effects of overall 24-hour glucocorticoid overexposure and the effects of a lack of circadian rhythmicity. For this, she continuously treated rats with a low dose of corticosterone (or "B"), which leads to a constant hormone level, without 24-hour overexposure using subcutaneously implanted pellets. The data from this "B-flat" model suggest that even modest elevations of glucocorticoid signaling during the time of the normal circadian trough of hormone secretion are a substantial contributor to the negative effects of glucocorticoids on health.


Asunto(s)
Glucocorticoides , Estrés Psicológico , Femenino , Ratas , Animales , Glucocorticoides/metabolismo , Corticosterona/metabolismo , Transducción de Señal , Ritmo Circadiano/fisiología , Receptores de Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo
5.
Eur J Neurosci ; 55(9-10): 2491-2518, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33724565

RESUMEN

Stressful experiences evoke, among others, a rapid increase in brain (nor)epinephrine (NE) levels and a slower increase in glucocorticoid hormones (GCs) in the brain. Microglia are key regulators of neuronal function and contain receptors for NE and GCs. These brain cells may therefore potentially be involved in modulating stress effects on neuronal function and learning and memory. In this review, we discuss that stress induces (1) an increase in microglial numbers as well as (2) a shift toward a pro-inflammatory profile. These microglia have (3) impaired crosstalk with neurons and (4) disrupted glutamate signaling. Moreover, microglial immune responses after stress (5) alter the kynurenine pathway through metabolites that impair glutamatergic transmission. All these effects could be involved in the impairments in memory and in synaptic plasticity caused by (prolonged) stress, implicating microglia as a potential novel target in stress-related memory impairments.


Asunto(s)
Microglía , Plasticidad Neuronal , Glucocorticoides/metabolismo , Humanos , Trastornos de la Memoria/metabolismo , Microglía/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo
6.
Eur J Neurosci ; 55(9-10): 2666-2683, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33840130

RESUMEN

Glucocorticoids enhance memory consolidation of emotionally arousing events via largely unknown molecular mechanisms. This glucocorticoid effect on the consolidation process also requires central noradrenergic neurotransmission. The intracellular pathways of these two stress mediators converge on two transcription factors: the glucocorticoid receptor (GR) and phosphorylated cAMP response element-binding protein (pCREB). We therefore investigated, in male rats, whether glucocorticoid effects on memory are associated with genomic interactions between the GR and pCREB in the hippocampus. In a two-by-two design, object exploration training or no training was combined with post-training administration of a memory-enhancing dose of corticosterone or vehicle. Genomic effects were studied by chromatin immunoprecipitation followed by sequencing (ChIP-seq) of GR and pCREB 45 min after training and transcriptome analysis after 3 hr. Corticosterone administration induced differential GR DNA-binding and regulation of target genes within the hippocampus, largely independent of training. Training alone did not result in long-term memory nor did it affect GR or pCREB DNA-binding and gene expression. No strong evidence was found for an interaction between GR and pCREB. Combination of the GR DNA-binding and transcriptome data identified a set of novel, likely direct, GR target genes that are candidate mediators of corticosterone effects on memory consolidation. Cell-specific expression of the identified target genes using single-cell expression data suggests that the effects of corticosterone reflect in part non-neuronal cells. Together, our data identified new GR targets associated with memory consolidation that reflect effects in both neuronal and non-neuronal cells.


Asunto(s)
Glucocorticoides , Consolidación de la Memoria , Animales , Corticosterona/metabolismo , Corticosterona/farmacología , ADN/metabolismo , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Hipocampo/metabolismo , Masculino , Ratas , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
7.
Ophthalmologica ; 245(6): 555-562, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35843207

RESUMEN

PURPOSE: The aim of the study was to assess the role of sex hormones in male and female patients with central serous chorioretinopathy (CSC), a disease with a pronounced male predilection. METHODS: A total of 206 chronic CSC patients (183 males, 23 females) and 59 healthy controls (29 males, 30 females) were enrolled. Serum testosterone, estradiol, albumin, and sex hormone-binding globulin levels were determined using immunoassays. The free fraction of testosterone and the free testosterone/estradiol ratio were calculated. RESULTS: No differences in the levels of total testosterone and estradiol were observed between CSC patients and healthy controls. Albumin levels were found to be lower in male CSC patients compared to controls (controls 47.8 g/L, patients 46.0 g/L, adj. p = 0.006). Only in females with CSC, sex hormone-binding globulin levels were found to be lower (controls 94.2 nmol/L, patients 50.4 nmol/L, adj. p = 0.001), together with a higher free testosterone/estradiol ratio (controls 0.06, patients 0.18, adj. p = 0.018). CONCLUSIONS: In this study, we did not find evidence for a disturbance in sex hormone levels in males with CSC. The lower levels of sex hormone-binding globulin in females with CSC, leading to a disturbed free testosterone/estradiol ratio, warrant further investigation into the role of androgens in females with CSC.


Asunto(s)
Coriorretinopatía Serosa Central , Globulina de Unión a Hormona Sexual , Humanos , Masculino , Femenino , Coriorretinopatía Serosa Central/diagnóstico , Hormonas Esteroides Gonadales , Testosterona , Estradiol , Albúminas
8.
Int J Mol Sci ; 24(1)2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36613751

RESUMEN

Angelman Syndrome (AS) is a severe neurodevelopmental disorder, caused by the neuronal absence of the ubiquitin protein ligase E3A (UBE3A). UBE3A promotes ubiquitin-mediated protein degradation and functions as a transcriptional coregulator of nuclear hormone receptors, including the glucocorticoid receptor (GR). Previous studies showed anxiety-like behavior and hippocampal-dependent memory disturbances in AS mouse models. Hippocampal GR is an important regulator of the stress response and memory formation, and we therefore investigated whether the absence of UBE3A in AS mice disrupted GR signaling in the hippocampus. We first established a strong cortisol-dependent interaction between the GR ligand binding domain and a UBE3A nuclear receptor box in a high-throughput interaction screen. In vivo, we found that UBE3A-deficient AS mice displayed significantly more variation in circulating corticosterone levels throughout the day compared to wildtypes (WT), with low to undetectable levels of corticosterone at the trough of the circadian cycle. Additionally, we observed an enhanced transcriptomic response in the AS hippocampus following acute corticosterone treatment. Surprisingly, chronic corticosterone treatment showed less contrast between AS and WT mice in the hippocampus and liver transcriptomic responses. This suggests that UBE3A limits the acute stimulation of GR signaling, likely as a member of the GR transcriptional complex. Altogether, these data indicate that AS mice are more sensitive to acute glucocorticoid exposure in the brain compared to WT mice. This suggests that stress responsiveness is altered in AS which could lead to anxiety symptoms.


Asunto(s)
Síndrome de Angelman , Ratones , Animales , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Corticosterona/metabolismo , Hipocampo/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Modelos Animales de Enfermedad
9.
Eur J Neurosci ; 54(7): 6374-6381, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34498316

RESUMEN

Neuroimaging studies suggest that intranasal oxytocin (IN-OXT) may modulate emotional and social processes by altering neural activity patterns. The extent of brain penetration after IN-OXT is unclear, and it is currently unknown whether IN-OXT can directly bind central oxytocin receptors (OXTRs). We investigated oxytocin pathway gene expression in regions affected by IN-OXT on task-based fMRI. We found that OXTR is more highly expressed in affected than unaffected subcortical regions; this effect did not vary by task type or sex. Cortical results revealed higher OXTR expression in regions affected by IN-OXT in emotional processing tasks and in male-only data. No significant differences were found in expression of the closely related vasopressin receptors. Our findings suggest that the mechanism by which IN-OXT may alter brain functionality involves direct activation of central OXTRs.


Asunto(s)
Oxitocina , Receptores de Oxitocina , Administración Intranasal , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo
10.
FASEB J ; 34(1): 1150-1168, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914623

RESUMEN

Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis occurs early in Alzheimer's disease (AD), associated with elevated circulating glucocorticoids (GC) and glucocorticoid receptors (GR) signaling impairment. However, the precise role of GR in the pathophysiology of AD remains unclear. Using an acute model of AD induced by the intracerebroventricular injection of amyloid-ß oligomers (oAß), we analyzed cellular and behavioral hallmarks of AD, GR signaling pathways, processing of amyloid precursor protein, and enzymes involved in Tau phosphorylation. We focused on the prefrontal cortex (PFC), particularly rich in GR, early altered in AD and involved in HPA axis control and cognitive functions. We found that oAß impaired cognitive and emotional behaviors, increased plasma GC levels, synaptic deficits, apoptosis and neuroinflammatory processes. Moreover, oAß potentiated the amyloidogenic pathway and enzymes involved both in Tau hyperphosphorylation and GR activation. Treatment with a selective GR modulator (sGRm) normalized plasma GC levels and all behavioral and biochemical parameters analyzed. GR seems to occupy a central position in the pathophysiology of AD. Deregulation of the HPA axis and a feed-forward effect on PFC GR sensitivity could participate in the etiology of AD, in perturbing Aß and Tau homeostasis. These results also reinforce the therapeutic potential of sGRm in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Receptores de Glucocorticoides/metabolismo , Proteínas tau/metabolismo , Corticoesteroides/química , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Conducta Animal , Modelos Animales de Enfermedad , Glucocorticoides/metabolismo , Homeostasis , Sistema Hipotálamo-Hipofisario , Masculino , Fosforilación , Sistema Hipófiso-Suprarrenal , Ratas , Ratas Sprague-Dawley , Transducción de Señal
11.
Stress ; 24(2): 130-153, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32755268

RESUMEN

The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.


Asunto(s)
Receptores de Glucocorticoides , Estrés Psicológico , Teorema de Bayes , Encéfalo/metabolismo , Glucocorticoides , Humanos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
12.
Pharmacol Res ; 168: 105588, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33798733

RESUMEN

Glucocorticoids regulate numerous processes in human physiology, but deregulated or excessive glucocorticoid receptor (GR) signaling contributes to the development of various pathologies including metabolic syndrome. For this reason, GR antagonists have considerable therapeutic value. Yet, the only GR antagonist that is clinically approved to date - mifepristone - exhibits cross-reactivity with other nuclear steroid receptors like the progesterone receptor. In this study, we set out to identify novel selective GR antagonists by combining rational chemical design with an unbiased in vitro and in vivo screening approach. Using this pipeline, we were able to identify CORT125329 as the compound with the best overall profile from our octahydro series of novel GR antagonists, and demonstrated that CORT125329 does not exhibit cross-reactivity with the progesterone receptor. Further in vivo testing showed beneficial activities of CORT125329 in models for excessive corticosterone exposure and short- and long-term high-fat diet-induced metabolic complications. Upon CORT125329 treatment, most metabolic parameters that deteriorated upon high-fat diet feeding were similarly improved in male and female mice, confirming activity in both sexes. However, some sexually dimorphic effects were observed including male-specific antagonism of GR activity in brown adipose tissue and female-specific lipid lowering activities after short-term CORT125329 treatment. Remarkably, CORT125329 exhibits beneficial metabolic effects despite its lack of GR antagonism in white adipose tissue. Rather, we propose that CORT125329 treatment restores metabolic activity in brown adipose tissue by stimulating lipolysis, mitochondrial activity and thermogenic capacity. In summary, we have identified CORT125329 as a selective GR antagonist with strong beneficial activities in metabolic disease models, paving the way for further clinical investigation.


Asunto(s)
Enfermedades Metabólicas/tratamiento farmacológico , Receptores de Glucocorticoides/antagonistas & inhibidores , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Dieta Alta en Grasa , Diseño de Fármacos , Desarrollo de Medicamentos , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Mol Cell Neurosci ; 108: 103541, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32858150

RESUMEN

The Single Prolonged Stress protocol is considered a model for PTSD, as it induces long lasting changes in rat behaviour and endocrine regulation. Previous work demonstrated that some of these changes can be prevented by treatment with the glucocorticoid receptor antagonist RU486, administered a week after the stressor. The current study evaluated the effects of an earlier intervention with RU486, as evaluated 1 week after SPS-exposure. Most RU486 effects occurred independent of prior stress, except for the reversal of a stress-induced increase in locomotor behaviour. The accompanying changes in gene expression depended on gene, brain region, and time. DNA methylation of the robustly down-regulated Fkbp5 gene was dissociated of changes in mRNA expression. The findings reinforce the long term effects of GR antagonist treatment, but also emphasize the need to evaluate changes over time to allow the identification of robust correlates between gene expression and behavioural/endocrine outcome of stressful experiences.


Asunto(s)
Antagonistas de Hormonas/uso terapéutico , Mifepristona/uso terapéutico , Trastornos por Estrés Postraumático/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Metilación de ADN , Esquema de Medicación , Antagonistas de Hormonas/administración & dosificación , Antagonistas de Hormonas/farmacología , Masculino , Mifepristona/administración & dosificación , Mifepristona/farmacología , Ratas , Ratas Wistar , Receptores de Glucocorticoides/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
14.
Front Neuroendocrinol ; 52: 206-218, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30590067

RESUMEN

In contrast to classical conditioning of physiological responses such as immune responses and drug effects, only a limited number of studies investigated classical conditioning of endocrine responses. The present paper is the first systematic review that integrates evidence from animal and human trials regarding the possibility to condition the endocrine responses. Twenty-six animal and eight human studies were included in the review. We demonstrated that there is accumulating evidence that classical conditioning processes are able to influence specific endocrine responses, such as cortocosterone/cortisol and insulin, while more limited evidence exists for other hormones. Animal and human studies were generally consistent in their findings; however, the limited number of human studies makes it difficult to generalize and translate the results of animal research to humans. Next to methodological recommendations for future studies, we suggest several ways how classically conditioned endocrine responses can be used in clinical practice.


Asunto(s)
Condicionamiento Clásico/fisiología , Corticosterona/metabolismo , Hidrocortisona/metabolismo , Insulina/metabolismo , Animales , Humanos
15.
Cell Mol Neurobiol ; 39(4): 539-549, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30291573

RESUMEN

Corticosteroid hormones act in the brain to support adaptation to stress via binding to mineralocorticoid and glucocorticoid receptors (MR and GR). These receptors act in large measure as transcription factors. Corticosteroid effects can be highly divergent, depending on the receptor type, but also on brain region, cell type, and physiological context. These differences ultimately depend on differential interactions of MR and GR with other proteins, which determine ligand binding, nuclear translocation, and transcriptional activities. In this review, we discuss established and potential mechanisms that confer receptor and cell type-specific effects of the MR and GR-mediated transcriptional effects in the brain.


Asunto(s)
Encéfalo/metabolismo , Receptores de Esteroides/metabolismo , Transcripción Genética , Animales , Genoma , Humanos , Modelos Biológicos , Receptores de Esteroides/química , Receptores de Esteroides/genética , Activación Transcripcional/genética
16.
Cell Mol Neurobiol ; 39(4): 503-522, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30173378

RESUMEN

Psychotic depression is characterized by elevated circulating cortisol, and high daily doses of the glucocorticoid/progesterone antagonist mifepristone for 1 week are required for significant improvement. Using a rodent model, we find that such high doses of mifepristone are needed because the antagonist is rapidly degraded and poorly penetrates the blood-brain barrier, but seems to facilitate the entry of cortisol. We also report that in male C57BL/6J mice, after a 7-day treatment with a high dose of mifepristone, basal blood corticosterone levels were similar to that of vehicle controls. This is surprising because after the first mifepristone challenge, corticosterone remained elevated for about 16 h, and then decreased towards vehicle control levels at 24 h. At that time, stress-induced corticosterone levels of the 1xMIF were sevenfold higher than the 7xMIF group, the latter response being twofold lower than controls. The 1xMIF mice showed behavioral hyperactivity during exploration of the circular hole board, while the 7xMIF mice rather engaged in serial search patterns. To explain this rapid reset of corticosterone secretion upon recurrent mifepristone administration, we suggest the following: (i) A rebound glucocorticoid feedback after cessation of mifepristone treatment. (ii) Glucocorticoid agonism in transrepression and recruitment of cell-specific coregulator cocktails. (iii) A more prominent role of brain MR function in control of stress circuit activity. An overview table of neuroendocrine MIF effects is provided. The data are of interest for understanding the mechanistic underpinning of stress system reset as treatment strategy for stress-related diseases.


Asunto(s)
Mifepristona/farmacología , Estrés Fisiológico/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Corticosterona/sangre , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Humanos , Hidrocortisona/metabolismo , Masculino , Ratones Endogámicos C57BL , Mifepristona/administración & dosificación , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo
17.
Neuroendocrinology ; 109(3): 266-276, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30884490

RESUMEN

Glucocorticoid hormones have important effects on brain function in the context of acute and chronic stress. Many of these are mediated by the glucocorticoid receptor (GR). GR has transcriptional activity which is highly context-specific and differs between tissues and even between cell types. The outcome of GR-mediated transcription depends on the interactome of associated coregulators. Selective GR modulators (SGRMs) are a class of GR ligands that can be used to activate only a subset of GR-coregulator interactions, thereby giving the possibility to induce a unique combination of agonistic and antagonistic GR properties. We describe SGRM action in animal models of brain function and pathology, and argue for their utility as molecular filters, to characterize context-specific GR interactome and transcriptional activity that are responsible for particular glucocorticoid-driven effects in cognitive processes such as memory consolidation. The ultimate objective of this approach is to identify molecular processes that are responsible for adaptive and maladaptive effects of glucocorticoids in the brain.


Asunto(s)
Encéfalo/metabolismo , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Humanos
18.
Proc Natl Acad Sci U S A ; 113(10): 2738-43, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26811448

RESUMEN

Steroid receptors are pleiotropic transcription factors that coordinate adaptation to different physiological states. An important target organ is the brain, but even though their effects are well studied in specific regions, brain-wide steroid receptor targets and mediators remain largely unknown due to the complexity of the brain. Here, we tested the idea that novel aspects of steroid action can be identified through spatial correlation of steroid receptors with genome-wide mRNA expression across different regions in the mouse brain. First, we observed significant coexpression of six nuclear receptors (NRs) [androgen receptor (Ar), estrogen receptor alpha (Esr1), estrogen receptor beta (Esr2), glucocorticoid receptor (Gr), mineralocorticoid receptor (Mr), and progesterone receptor (Pgr)] with sets of steroid target genes that were identified in single brain regions. These coexpression relationships were also present in distinct other brain regions, suggestive of as yet unidentified coordinate regulation of brain regions by, for example, glucocorticoids and estrogens. Second, coexpression of a set of 62 known NR coregulators and the six steroid receptors in 12 nonoverlapping mouse brain regions revealed selective downstream pathways, such as Pak6 as a mediator for the effects of Ar and Gr on dopaminergic transmission. Third, Magel2 and Irs4 were identified and validated as strongly responsive targets to the estrogen diethylstilbestrol in the mouse hypothalamus. The brain- and genome-wide correlations of mRNA expression levels of six steroid receptors that we provide constitute a rich resource for further predictions and understanding of brain modulation by steroid hormones.


Asunto(s)
Encéfalo/metabolismo , Perfilación de la Expresión Génica/métodos , Genoma/genética , Receptores de Esteroides/genética , Transducción de Señal/genética , Animales , Receptor alfa de Estrógeno/genética , Hipocampo/metabolismo , Hibridación in Situ , Hibridación Fluorescente in Situ , Masculino , Ratones Endogámicos C57BL , Receptores de Progesterona/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Int J Mol Sci ; 20(7)2019 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-30934833

RESUMEN

Mineralocorticoid receptor (MR)-mediated signaling in the brain has been suggested as a protective factor in the development of psychopathology, in particular mood disorders. We recently identified genomic loci at which either MR or the closely related glucocorticoid receptor (GR) binds selectively, and found members of the NeuroD transcription factor family to be specifically associated with MR-bound DNA in the rat hippocampus. We show here using forebrain-specific MR knockout mice that GR binding to MR/GR joint target loci is not affected in any major way in the absence of MR. Neurod2 binding was also independent of MR binding. Moreover, functional comparison with MyoD family members indicates that it is the chromatin remodeling aspect of NeuroD, rather than its direct stimulation of transcription, that is responsible for potentiation of MR-mediated transcription. These findings suggest that NeuroD acts in a permissive way to enhance MR-mediated transcription, and they argue against competition for DNA binding as a mechanism of MR- over GR-specific binding.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Corticosterona/metabolismo , ADN/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Ratones Noqueados , Modelos Biológicos , Proteína MioD/metabolismo , Proteínas del Tejido Nervioso/química , Unión Proteica , Dominios Proteicos , Receptores de Glucocorticoides/metabolismo , Relación Estructura-Actividad
20.
Gut ; 67(7): 1269-1279, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29101261

RESUMEN

OBJECTIVE: Butyrate exerts metabolic benefits in mice and humans, the underlying mechanisms being still unclear. We aimed to investigate the effect of butyrate on appetite and energy expenditure, and to what extent these two components contribute to the beneficial metabolic effects of butyrate. DESIGN: Acute effects of butyrate on appetite and its method of action were investigated in mice following an intragastric gavage or intravenous injection of butyrate. To study the contribution of satiety to the metabolic benefits of butyrate, mice were fed a high-fat diet with butyrate, and an additional pair-fed group was included. Mechanistic involvement of the gut-brain neural circuit was investigated in vagotomised mice. RESULTS: Acute oral, but not intravenous, butyrate administration decreased food intake, suppressed the activity of orexigenic neurons that express neuropeptide Y in the hypothalamus, and decreased neuronal activity within the nucleus tractus solitarius and dorsal vagal complex in the brainstem. Chronic butyrate supplementation prevented diet-induced obesity, hyperinsulinaemia, hypertriglyceridaemia and hepatic steatosis, largely attributed to a reduction in food intake. Butyrate also modestly promoted fat oxidation and activated brown adipose tissue (BAT), evident from increased utilisation of plasma triglyceride-derived fatty acids. This effect was not due to the reduced food intake, but explained by an increased sympathetic outflow to BAT. Subdiaphragmatic vagotomy abolished the effects of butyrate on food intake as well as the stimulation of metabolic activity in BAT. CONCLUSION: Butyrate acts on the gut-brain neural circuit to improve energy metabolism via reducing energy intake and enhancing fat oxidation by activating BAT.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Apetito/efectos de los fármacos , Butiratos/farmacología , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Administración Oral , Animales , Butiratos/administración & dosificación , Inyecciones Intravenosas , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA