Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 479: 91-106, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34762981

RESUMO

Like other members of the superfamily of nuclear receptors, the peroxisome proliferator-activated receptor γ (PPARγ), is a ligand-activated transcription factor known for its insulin-sensitizing actions in the periphery. Despite only sparse evidence for PPARγ in the CNS, many reports suggest direct PPARγ-mediated actions in the brain. This study aimed to (i) map PPARγ expression in rodent brain areas, involved in the regulation of cognitive, motivational, and emotional functions, (ii) examine the regulation of central PPARγ by physiological variables (age, sex, obesity); (iii) chemotypically identify PPARγ-expressing cells in the frontal cortex (FC) and hippocampus (HP); (iv) study whether activation of PPARγ by pioglitazone (Pio) in FC and HP cells can induce target gene expression; and (v) demonstrate the impact of activated PPARγ on learning behavior and motivation. Immunoreactive PPARγ was detectable in specific sub-nuclei/subfields of the FC, HP, nucleus accumbens, amygdala, hypothalamus, thalamus, and granular layers of the cerebellum. PPARγ protein levels were upregulated during aging and in high fat diet-induced obesity. PPARγ mRNA expression was upregulated in the amygdala of females (but not males) that were made obese. Neural precursor cells, mature neurons, and astrocytes in primary FC and HP cultures were shown to express PPARγ. Pioglitazone dose-dependently upregulated PPARγ target genes in manner that was specific to the origin (FC or HP) of the cultures. Lastly, administration of Pio impaired motivation and associative learning. Collectively, we provide evidence for the presence of regulatable PPARγ in the brain and demonstrate their participation the regulation of key behaviors.


Assuntos
Células-Tronco Neurais , Tiazolidinedionas , Encéfalo/metabolismo , Feminino , Humanos , Masculino , Motivação , Células-Tronco Neurais/metabolismo , PPAR gama/metabolismo , Pioglitazona/farmacologia , Tiazolidinedionas/farmacologia
2.
Acta Physiol (Oxf) ; 228(2): e13345, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31310704

RESUMO

AIM: Since foods with high hedonic value are often consumed in excess of energetic needs, this study was designed to identify the mechanisms that may counter anorexigenic signalling in the presence of hedonic foods in lean animals. METHODS: Mice, in different states of satiety (fed/fasted, or fed/fasted and treated with ghrelin or leptin, respectively), were allowed to choose between high-fat/high-sucrose and standard foods. Intake of each food type and the activity of hypothalamic neuropetidergic neurons that regulate appetite were monitored. In some cases, food choice was monitored in leptin-injected fasted mice that received microinjections of galanin receptor agonists into the lateral hypothalamus. RESULTS: Appetite-stimulating orexin neurons in the lateral hypothalamus are rapidly activated when lean, satiated mice consume a highly palatable food (PF); such activation (upregulated c-Fos expression) occurred even after administration of the anorexigenic hormone leptin and despite intact leptin signalling in the hypothalamus. The ability of leptin to restrain PF eating is restored when a galanin receptor 2 (Gal2R) agonist is injected into the lateral hypothalamus. CONCLUSION: Hedonically-loaded foods interrupt the inhibitory actions of leptin on orexin neurons and interfere with the homeostatic control of feeding. Overeating of palatable foods can be curtailed in lean animals by activating Gal2R in the lateral hypothalamus.


Assuntos
Ingestão de Alimentos/fisiologia , Hiperfagia/prevenção & controle , Região Hipotalâmica Lateral/efeitos dos fármacos , Leptina/farmacologia , Neurônios/metabolismo , Receptor Tipo 2 de Galanina/agonistas , Animais , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Galanina/farmacologia , Grelina/metabolismo , Hiperfagia/metabolismo , Hiperfagia/patologia , Região Hipotalâmica Lateral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Orexinas/metabolismo , Receptor Tipo 2 de Galanina/metabolismo
3.
J Neurosci ; 38(2): 441-451, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29196316

RESUMO

Early-life obesity predisposes to obesity in adulthood, a condition with broad medical implications including sleep disorders, which can exacerbate metabolic disturbances and disrupt cognitive and affective behaviors. In this study, we examined the long-term impact of transient peripubertal diet-induced obesity (ppDIO, induced between 4 and 10 weeks of age) on sleep-wake behavior in male mice. EEG and EMG recordings revealed that ppDIO increases sleep during the active phase but reduces resting-phase sleep quality. This impaired sleep phenotype persisted for up to 1 year, although animals were returned to a non-obesiogenic diet from postnatal week 11 onwards. To better understand the mechanisms responsible for the ppDIO-induced alterations in sleep, we focused on the lateral hypothalamus (LH). Mice exposed to ppDIO did not show altered mRNA expression levels of orexin and melanin-concentrating hormone, two peptides that are important for sleep-wake behavior and food intake. Conversely, the LH of ppDIO-exposed mice had reduced contents of serotonin (5-hydroxytryptamine, 5-HT), a neurotransmitter involved in both sleep-wake and satiety regulation. Interestingly, an acute peripheral injection of the satiety-signaling peptide YY 3-36 increased 5-HT turnover in the LH and ameliorated the ppDIO-induced sleep disturbances, suggesting the therapeutic potential of this peptide. These findings provide new insights into how sleep-wake behavior is programmed during early life and how peripheral and central signals are integrated to coordinate sleep.SIGNIFICANCE STATEMENT Adult physiology and behavior are strongly influenced by dynamic reorganization of the brain during puberty. The present work shows that obesity during puberty leads to persistently dysregulated patterns of sleep and wakefulness by blunting serotonergic signaling in the lateral hypothalamus. It also shows that pharmacological mimicry of satiety with peptide YY3-36 can reverse this neurochemical imbalance and acutely restore sleep composition. These findings add insight into how innate behaviors such as feeding and sleep are integrated and suggest a novel mechanism through which diet-induced obesity during puberty imposes its long-lasting effects on sleep-wake behavior.


Assuntos
Região Hipotalâmica Lateral/metabolismo , Obesidade/complicações , Serotonina/metabolismo , Transtornos do Sono-Vigília/etiologia , Animais , Homeostase/fisiologia , Região Hipotalâmica Lateral/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Transtornos do Sono-Vigília/metabolismo
4.
Epigenetics ; 10(3): 247-57, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793778

RESUMO

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.


Assuntos
Metilação de DNA , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/metabolismo , Animais , Células Cultivadas , Ilhas de CpG , Hipotálamo/metabolismo , Masculino , Camundongos Endogâmicos C57BL
5.
J Comp Physiol B ; 184(8): 1055-63, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25179180

RESUMO

The wild sand rat, Psammomys obesus, displays seasonal variations in adrenocortical activity that parallel those of testicular activity, indicating functional cross-talk between the hypothalamo-pituitary-adrenal and hypothalamo-pituitary-gonadal axes. In the present study, we examined androgen receptor (AR)-mediated actions of testicular steroids in the regulation of adrenocortical function in the sand rat. Specifically, we examined the expression of AR in the adrenal cortex, as well as adrenal apoptosis in male sand rats that had been surgically castrated or castrated and supplemented with testosterone; biochemical indices of adrenocortical function and hormone profiles were also measured. Orchiectomy was followed by an increase in adrenocorticotropic hormone secretion from the anterior pituitary and subsequently, increased adrenocortical activity; the latter was evidenced by orchiectomy-induced increases in the adrenal content of cholesterol and lipids as well as adrenal hypertrophy (seen as an elevation of the RNA/DNA ratio). Further, androgen deprivation respectively up- and downregulated the incidence of apoptosis within the glucocorticoid-producing zona fasciculata and sex steroid-producing zona reticularis. Interestingly, orchiectomy resulted in increased expression of AR in the zona fasciculata. All of the orchiectomy-induced cellular and biochemical responses were reversible after testosterone substitution therapy. Together, these data suggest that adrenocortical activity in the sand rat is seasonally modulated by testicular androgens that act through AR located in the adrenal cortex itself.


Assuntos
Adaptação Biológica/fisiologia , Córtex Suprarrenal/metabolismo , Gerbillinae/fisiologia , Hormônios Esteroides Gonadais/metabolismo , Receptores Androgênicos/metabolismo , Reprodução/fisiologia , Estações do Ano , Córtex Suprarrenal/citologia , Análise de Variância , Animais , Apoptose/fisiologia , Colesterol/metabolismo , Gerbillinae/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Metabolismo dos Lipídeos/fisiologia , Masculino , Orquiectomia , Testosterona/administração & dosagem , Testosterona/farmacologia
6.
Endocrinology ; 155(5): 1751-62, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24506071

RESUMO

Early-life stress (ELS) increases the vulnerability thresholds for stress-related diseases such as major depression and anxiety by inducing alterations in the structure and function of neural circuits and endocrine pathways. We previously demonstrated the contribution of epigenetic mechanisms to the long-term programming of the hypothalamo-pituitary-adrenal axis activity following ELS exposure in male mice. Here, ELS comprising daily separation of pups from their dams on postnatal days 1-10 was observed to up-regulate the expression of the pituitary proopiomelanocortin (Pomc) gene; POMC serves as a prohormone for ACTH, a key mediator of the adrenocortical response to stress. Detailed analysis revealed that the increase in Pomc mRNA levels results from a reduction in DNA methylation at a critical regulatory region of the Pomc gene; interestingly, this change occurs with some delay after ELS and persists for up to 1 year. Using a Pomc-expressing pituitary cell line (AtT20), we confirmed a role for DNA methylation in restraining Pomc expression under resting conditions: specifically, we show that CpG site-specific methylation of the Pomc promoter represses Pomc mRNA transcription. Further, we show high-affinity binding of methyl-CpG binding protein-2 to the distal promoter of Pomc, suggesting that methyl-CpG binding protein-2 acts in association with the chromatin modifiers histone deacetylase 2 and DNA methyltransferase 1 to repress Pomc gene expression. Collectively, these experiments contribute to our understanding of the mechanisms through which environmental cues are translated into stable changes ("cellular memory") in neuroendocrine cells.


Assuntos
Metilação de DNA , Modelos Animais de Doenças , Regulação para Baixo , Epigênese Genética , Hipófise/metabolismo , Pró-Opiomelanocortina/metabolismo , Estresse Psicológico/metabolismo , Animais , Linhagem Celular , Cinética , Masculino , Privação Materna , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Células Neuroendócrinas/metabolismo , Neurogênese , Neurônios/metabolismo , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Estresse Psicológico/etiologia , Transcrição Gênica
7.
J Biol Chem ; 288(33): 23725-39, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23818519

RESUMO

Despite numerous descriptions of rapid effects of corticosterone on neuronal function, the intracellular mechanisms responsible for these changes remain elusive. The present comprehensive analysis reveals that signaling from a membrane-located G protein-coupled receptor activates PKC, Akt/PKB, and PKA, which subsequently trigger the phosphorylation of the tyrosine kinases Pyk2, Src, and Abl. These changes induce rapid cytoskeletal rearrangements (increased PSD-95 co-clustering) within the post-synaptic density; these events are accompanied by increased surface NMDA receptor expression, reflecting corticosterone-induced inhibition of NMDA receptor endocytosis. Notably, none of these signaling mechanisms require de novo protein synthesis. The observed up-regulation of ERK1/2 (downstream of NMDA receptor signaling) together with the fact that c-Abl integrates cytoplasmic and nuclear functions introduces a potential mechanism through which rapid signaling initiated at the plasma membrane may eventually determine the long term integrated response to corticosterone by impacting on the transcriptional machinery that is regulated by classical, nuclear mineralocorticoid, and glucocorticoid receptors.


Assuntos
Glucocorticoides/metabolismo , Hipocampo/citologia , Neurônios/enzimologia , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Actinas/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Corticosterona/farmacologia , Ativação Enzimática/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Modelos Biológicos , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Mol Brain ; 3: 2, 2010 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-20180948

RESUMO

Studies into the mechanisms of corticosteroid action continue to be a rich bed of research, spanning the fields of neuroscience and endocrinology through to immunology and metabolism. However, the vast literature generated, in particular with respect to corticosteroid actions in the brain, tends to be contentious, with some aspects suffering from loose definitions, poorly-defined models, and appropriate dissection kits. Here, rather than presenting a comprehensive review of the subject, we aim to present a critique of key concepts that have emerged over the years so as to stimulate new thoughts in the field by identifying apparent shortcomings. This article will draw on experience and knowledge derived from studies of the neural actions of other steroid hormones, in particular estrogens, not only because there are many parallels but also because 'learning from differences' can be a fruitful approach. The core purpose of this review is to consider the mechanisms through which corticosteroids might act rapidly to alter neural signaling.


Assuntos
Corticosteroides , Encéfalo , Corticosteroides/metabolismo , Corticosteroides/farmacologia , Animais , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Neurônios/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Sinapses/fisiologia
9.
Ann Neurol ; 67(1): 21-30, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20186952

RESUMO

OBJECTIVE: Glucocorticoids (GCs) are indicated for a number of conditions in obstetrics and perinatal medicine; however, the neurodevelopmental and long-term neurological consequences of early-life GC exposure are still largely unknown. Preclinical studies have demonstrated that GCs have a major influence on hippocampal cell turnover by inhibiting neurogenesis and stimulating apoptosis of mature neurons. Here we examined the fate of the limited pool of neural progenitor cells (NPCs) after GC administration during neonatal development; the impact of this treatment on hippocampal structure was also studied. METHODS: Phenotype-specific genetic and antigenic markers were used to identify cultured NPCs at various developmental stages; the survival of these cells was monitored after exposure to the synthetic glucocorticoid dexamethasone (DEX). In addition, the effects of neonatal DEX treatment on the neurogenic potential of the rat hippocampus were examined by monitoring the incorporation of bromodeoxyuridine and expression of Ki67 antigen at various postnatal ages. RESULTS: Multipotent nestin-expressing NPCs and Talpha1-tubulin-expressing immature neurons succumb to GC-induced apoptosis in primary hippocampal cultures. Neonatal GC treatment results in marked apoptosis among the proliferating population of cells in the dentate gyrus, depletes the NPC pool, and leads to significant and sustained reductions in the volume of the dentate gyrus. INTERPRETATION: Both NPCs and immature neurons in the hippocampus are sensitive to the proapoptotic actions of GCs. Depletion of the limited NPC pool during early life retards hippocampal growth, thus allowing predictions about the potential neurological and psychiatric consequences of neonatal GC exposure.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Giro Denteado/efeitos dos fármacos , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/fisiologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Proteínas de Filamentos Intermediários/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/fisiologia , Ratos , Ratos Wistar , Nicho de Células-Tronco/crescimento & desenvolvimento , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Tubulina (Proteína)/metabolismo
10.
Nat Neurosci ; 12(12): 1559-66, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19898468

RESUMO

Adverse early life events can induce long-lasting changes in physiology and behavior. We found that early-life stress (ELS) in mice caused enduring hypersecretion of corticosterone and alterations in passive stress coping and memory. This phenotype was accompanied by a persistent increase in arginine vasopressin (AVP) expression in neurons of the hypothalamic paraventricular nucleus and was reversed by an AVP receptor antagonist. Altered Avp expression was associated with sustained DNA hypomethylation of an important regulatory region that resisted age-related drifts in methylation and centered on those CpG residues that serve as DNA-binding sites for the methyl CpG-binding protein 2 (MeCP2). We found that neuronal activity controlled the ability of MeCP2 to regulate activity-dependent transcription of the Avp gene and induced epigenetic marking. Thus, ELS can dynamically control DNA methylation in postmitotic neurons to generate stable changes in Avp expression that trigger neuroendocrine and behavioral alterations that are frequent features in depression.


Assuntos
Metilação de DNA/fisiologia , Depressão/fisiopatologia , Epigênese Genética/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Estresse Fisiológico/genética , Animais , Animais Recém-Nascidos , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Comportamento Animal/fisiologia , Corticosterona/sangue , Depressão/genética , Elementos Facilitadores Genéticos/fisiologia , Sistema Hipotálamo-Hipofisário/citologia , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Fenótipo , Fosforilação/genética , Transcrição Gênica/fisiologia
11.
Semin Fetal Neonatal Med ; 14(3): 130-5, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19084485

RESUMO

Epidemiological evidence links exposure to stress hormones during fetal or early postnatal development with lifetime prevalence of cardiac, metabolic, auto-immune, neurological and psychiatric disorders. This has led to the concept of 'developmental programming through stress'. Importantly, these effects (specifically, hypertension, hyperglycaemia and neurodevelopmental and behavioural abnormalities) can be reproduced by exposure to high glucocorticoid levels, indicating a crucial role of glucocorticoids in their causation. However, there can be important differences in outcome, depending on the exact time of exposure, as well as duration and receptor selectivity of the glucocorticoid applied. The mechanisms underlying programming by stress are still unclear but it appears that these environmental perturbations exploit epigenetic modifications of DNA and/or histones to induce stable modifications of gene expression. Programming of neuro- and behavioural development by glucocorticoids and stress are important determinants of lifetime health and should be a consideration when choosing treatments in obstetric and neonatal medicine.


Assuntos
Corticosteroides/fisiologia , Encéfalo/embriologia , Desenvolvimento Infantil/fisiologia , Animais , Feminino , Feto/fisiologia , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Lactente , Sistema Hipófise-Suprarrenal/fisiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Receptores de Glucocorticoides/fisiologia , Receptores de Mineralocorticoides/fisiologia , Estresse Fisiológico/fisiologia , Estresse Psicológico/fisiopatologia
12.
FASEB J ; 21(1): 231-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17135362

RESUMO

The transcription-intermediary-factor-2 (TIF-2) is a coactivator of the glucocorticoid receptor (GR), and its disruption would be expected to influence glucocorticoid-mediated control of the hypothalamo-pituitary-adrenal (HPA) axis. Here, we show that its targeted deletion in mice is associated with altered expression of several glucocorticoid-dependent components of HPA regulation (e.g., corticotropin-releasing hormone, vasopressin, ACTH, glucocorticoid receptors), suggestive of hyperactivity under basal conditions. At the same time, TIF-2(-/-) mice display significantly lower basal corticosterone levels and a sluggish and blunted initial secretory response to brief emotional and prolonged physical stress. Subsequent analysis revealed this discrepancy to result from pronounced aberrations in the structure and function of the adrenal gland, including the cytoarchitectural organization of the zona fasciculata and basal and stress-induced expression of key elements of steroid hormone synthesis, such as the steroidogenic acute regulatory (StAR) protein and 3beta-hydroxysteroid dehydrogenase (3beta-HSD). In addition, altered expression levels of two nuclear receptors, DAX-1 and steroidogenic factor 1 (SF-1), in the adrenal cortex strengthen the view that TIF-2 deletion disrupts adrenocortical development and steroid biosynthesis. Thus, hyperactivity of the hypothalamo-pituitary unit is ascribed to insidious adrenal insufficiency and impaired glucocorticoid feedback.


Assuntos
Córtex Suprarrenal/fisiopatologia , Receptores de Glucocorticoides/fisiologia , Fatores de Transcrição/fisiologia , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Sequência de Bases , Corticosterona/sangue , Primers do DNA , Feminino , Sistema Hipotálamo-Hipofisário , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coativador 2 de Receptor Nuclear , Fosfoproteínas/metabolismo , Sistema Hipófise-Suprarrenal , Receptores de Glucocorticoides/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Esteroidogênico 1 , Fatores de Transcrição/genética
13.
Dialogues Clin Neurosci ; 8(4): 417-32, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17290800

RESUMO

Illustrating the complexity of the stress response and its multifaceted manifestations is the leading idea of this overview of experimental paradigms used for stress induction in laboratory animals. The description of key features of models based on naturalistic stressors, pharmacological challenges, and genomic manipulations is complemented by comprehensive analysis of physiological, behavioral, neurochemical, and endocrine changes and their appropriateness as outcome readouts. Particular attention has been paid to the role of sex and age as determinants of the dynamics of the stress response. Possible translational applications of stress-inducing paradigms as models of disease are briefly sketched.


Assuntos
Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia , Envelhecimento/fisiologia , Envelhecimento/psicologia , Animais , Comportamento Animal , Modelos Animais de Doenças , Determinação de Ponto Final , Feminino , Humanos , Masculino , Caracteres Sexuais , Estresse Psicológico/imunologia , Estresse Psicológico/metabolismo
14.
Exp Gerontol ; 39(11-12): 1651-60, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15582281

RESUMO

The importance of androgen signaling is well recognized for numerous aspects of central nervous system (CNS) function, ranging from sex-specific organization of neuroendocrine and behavioral circuits to adaptive capacity, resistance and repair. Nonetheless, concepts for the therapeutic use of androgens in neurological and mental disorders are far from being established. This review outlines some critical issues which interfere with decisions on the suitability of androgens as therapeutic agents for CNS conditions. Among these, sex-specific organization of neural substrates and resulting differential responsiveness to endogenous gonadal steroids, convergence of steroid hormone actions on common molecular targets, co-presence of different sex steroid receptors in target neuronal populations, and in situ biotransformation of natural androgens apparently pose the principal obstacles for the characterization of specific neurotropic effects of androgens. Additional important, albeit less explored aspects consist in insufficient knowledge about molecular targets in the CNS which are under exclusive or predominant androgen control. Own experimental data illustrate the variability of pharmacological effects of natural and synthetic androgens on CNS functions of adaptive relevance, such as sexual behavior, anxiety and endocrine responsiveness to stress. Finally, we present results from an analysis of the consequences of aging for the rat brain transcriptome and examination of the influence of androgens on differentially expressed genes with presumable significance in neuropathology.


Assuntos
Androgênios/fisiologia , Encéfalo/metabolismo , Fatores de Crescimento Neural/fisiologia , Transdução de Sinais/fisiologia , Idoso , Envelhecimento , Androgênios/farmacologia , Animais , Humanos , Masculino , Modelos Animais , Ratos , Receptores Androgênicos/metabolismo , Comportamento Sexual , Estresse Psicológico
15.
FASEB J ; 18(13): 1568-70, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15289439

RESUMO

Transient activation of estrogen receptors (ER) in the developing brain during a limited perinatal "window of time" is recognized as a key mechanism of defeminization of neural control of reproductive function and sexual behavior. Two major ER isoforms, alpha and beta, are present in neural circuits that govern ovarian cycle and sexual behavior. Using highly selective ER agonists, this study provides the first evidence for distinct contribution of individual ER isoforms to the process of estrogen dependent defeminization. Neonatal activation of the ERalpha in female rats resulted in abrogation of cyclic ovarian activity and female sexual behavior in adulthood. These effects are associated with male-like alterations in the morphology of the anteroventral periventricular (AVPV) and sexually dimorphic nucleus of the preoptic area (SDN-POA), as well as refractoriness to estrogen-mediated induction of sexual receptivity. Exposure to an ERbeta-selective agonist induced persistent estrus and had a strong defeminizing effect on the hypothalamic gonadotropin "surge generator" AVPV. However, neonatal ERbeta activation failed to alter female sexual behavior, responsiveness to estrogens and morphometric features of the behaviorally relevant SDN-POA. Thus, although co-present in several brain regions involved in the control of female reproductive function, ER isoforms convey different, and probably not synergistic, chemical signals in the course of neonatal sex-specific brain organization.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Feminino , Ovário/efeitos dos fármacos , Ovário/fisiologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Ratos , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA