Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Alcohol Clin Exp Res ; 39(4): 631-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25752869

RESUMO

BACKGROUND: The clinical course of individuals exposed to alcohol in utero is influenced by multiple factors, including the social environments of the gravid female and offspring. In the present studies we focused on the effects of prenatal alcohol exposure (PAE) and the prenatal and early-life social environments on the hippocampal formation, as impaired development and functioning of this brain region have been implicated in several of the adverse cognitive outcomes associated with PAE. METHODS: We combined our PAE mouse model with 2 conditions of housing pregnant dams and their preweanling offspring: the standard nest (SN), in which a dam is individually housed prior to parturition and then remains isolated with her offspring, and the communal nest (CN), in which multiple dams are housed together prior to parturition and then following delivery the moms and their litters share a nest. Mouse dams consumed either 10% (w/v) ethanol in 0.066% (w/v) saccharin (SAC) or 0.066% (w/v) SAC alone using a limited (4-hour) access, drinking-in-the-dark paradigm. Immunoblotting techniques were used to measure levels of the glucocorticoid receptor (GR), 11-ß-hydroxysteroid dehydrogenase 1, the FK506-binding proteins 51 and 52, and corticotropin-releasing hormone receptor type 1 in the hippocampal formation isolated from male adolescent offspring. We also determined the effect of PAE and rearing conditions on context discrimination, a hippocampal-dependent learning/memory task. RESULTS: SN PAE offspring displayed impaired context discrimination and neurochemical changes in the hippocampal formation consistent with increased GR nuclear localization. These effects of PAE were, in general, ameliorated in mice reared in a CN. The CN also altered neurochemical measures and improved learning/memory in SAC control animals. CONCLUSIONS: These studies demonstrate a complex interplay between the effects of PAE and social environments. The findings have important translational implications, as well as highlight the importance of considering rearing conditions in the interpretation of research findings on PAE.


Assuntos
Etanol/efeitos adversos , Hipocampo/efeitos dos fármacos , Abrigo para Animais , Efeitos Tardios da Exposição Pré-Natal/psicologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Animais , Discriminação Psicológica/efeitos dos fármacos , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo
2.
Toxicol Rep ; 1: 544-553, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25243109

RESUMO

Fetal alcohol spectrum disorders (FASDs) are the number one cause of preventable mental retardation. An estimated 2-5% of children are diagnosed as having a FASD. While it is known that children prenatally exposed to alcohol experience cognitive deficits and a higher incidence of psychiatric illness later in life, the pathways underlying these abnormalities remain uncertain. GSK3ß and CDK5 are protein kinases that are converging points for a vast number of signaling cascades, including those controlling cellular processes critical to learning and memory. We investigated whether levels of GSK3ß and CDK5 are affected by moderate prenatal alcohol exposure (PAE), specifically in the hippocampus and medial frontal cortex of the adolescent mouse. In the present work we utilized immunoblotting techniques to demonstrate that moderate PAE increased hippocampal p35 and ß-catenin, and decreased total levels of GSK3ß, while increasing GSK3ß Ser9 and Tyr216 phosphorylation. Interestingly, different alterations were seen in the medial frontal cortex where p35 and CDK5 were decreased and increased total GSK3ß was accompanied by reduced Tyr216 of the enzyme. These results suggest that kinase dysregulation during adolescence might be an important contributing factor to the effects of PAE on hippocampal and medial frontal cortical functioning; and by extension, that global modulation of these kinases may produce differing effects depending on brain region.

3.
PLoS One ; 9(4): e96200, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24755652

RESUMO

Prenatal alcohol exposure (PAE) has been shown to impair learning, memory and executive functioning in children. Perseveration, or the failure to respond adaptively to changing contingencies, is a hallmark on neurobehavioral assessment tasks for human fetal alcohol spectrum disorder (FASD). Adaptive responding is predominantly a product of the medial prefrontal cortex (mPFC) and is regulated by corticosteroids. In our mouse model of PAE we recently reported deficits in hippocampal formation-dependent learning and memory and a dysregulation of hippocampal formation glucocorticoid receptor (GR) subcellular distribution. Here, we examined the effect of PAE on frontal cortical-dependent behavior, as well as mPFC GR subcellular distribution and the levels of regulators of intracellular GR transport. PAE mice displayed significantly reduced response flexibility in a Y-maze reversal learning task. While the levels of total nuclear GR were reduced in PAE mPFC, levels of GR phosphorylated at serines 203, 211 and 226 were not significantly changed. Cytosolic, but not nuclear, MR levels were elevated in the PAE mPFC. The levels of critical GR trafficking proteins, FKBP51, Hsp90, cyclophilin 40, dynamitin and dynein intermediate chain, were altered in PAE mice, in favor of the exclusion of GR from the nucleus, indicating dysregulation of GR trafficking. Our findings suggest that there may be a link between a deficit in GR nuclear localization and frontal cortical learning deficits in prenatal alcohol-exposed mice.


Assuntos
Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/metabolismo , Córtex Pré-Frontal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Glucocorticoides/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Membrana Celular/metabolismo , Peptidil-Prolil Isomerase F , Ciclofilinas/metabolismo , Feminino , Transtornos do Espectro Alcoólico Fetal/psicologia , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/psicologia , Transporte Proteico , Receptores de Mineralocorticoides/metabolismo , Reversão de Aprendizagem/efeitos dos fármacos , Proteínas de Ligação a Tacrolimo/metabolismo
4.
Alcohol Clin Exp Res ; 38(2): 392-400, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23992407

RESUMO

BACKGROUND: Accumulating evidence indicates that several of the long-term consequences of prenatal alcohol exposure (PAE) are the result of changes in the development and function of cortico-limbic structures, including the hippocampal formation. The glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) are key regulators of hippocampal formation development, structure, and functioning and, thus, are potential mediators of PAE's effects on this brain region. In the present studies, we assessed the impact of PAE on components of corticosteroid signaling pathways in the mouse hippocampal formation. METHODS: Throughout pregnancy, mouse dams were offered either 10% (w/v) ethanol sweetened with 0.066% (w/v) saccharin (SAC) or 0.066% (w/v) SAC alone using a limited (4-hour) access, drinking-in-the-dark paradigm. The hippocampal formation was isolated from naïve postnatal day 40 to 50 offspring, and subcellular fractions were prepared. Using immunoblotting techniques, we measured the levels of GR, MR, 11-ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1), and the FK506-binding proteins 51 (FKBP51, FKBP5) and 52 (FKBP52, FKBP4). Finally, we determined the effect of PAE on context discrimination, a hippocampal-dependent learning/memory task. RESULTS: PAE was associated with reduced MR and elevated GR nuclear localization in the hippocampal formation, whereas cytosolic levels of both receptors were not significantly altered. FKBP51 levels were reduced, while FKBP52 levels were unaltered, and 11ß-HSD1 levels were increased in postnuclear fractions isolated from PAE mouse hippocampal formation. These neurochemical alterations were associated with reduced context discrimination. CONCLUSIONS: The data support a model in which PAE leads to increased nuclear localization of GRs secondary to reductions in FKBP51 and increases in 11ß-HSD1 levels in the adolescent mouse hippocampal formation. Persistent dysregulation of GR subcellular distribution is predicted to damage the hippocampal formation and may underlie many of the effects of PAE on hippocampal-dependent functioning.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Hipocampo/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Animais , Western Blotting , Hormônio Liberador da Corticotropina/metabolismo , Interpretação Estatística de Dados , Discriminação Psicológica/efeitos dos fármacos , Feminino , Hipocampo/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos , Gravidez , Desempenho Psicomotor/efeitos dos fármacos , Frações Subcelulares/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo
5.
Neurotoxicology ; 33(5): 1338-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22960421

RESUMO

Over the past two decades, key advancements have been made in understanding the complex pathology that occurs following not only high levels of arsenic exposure (>1 ppm) but also levels previously considered to be low (<100 ppb). Past studies have characterized the deleterious effects of arsenic on the various functions of cardiovascular, pulmonary, immunological, respiratory, endocrine and neurological systems. Other research has demonstrated an elevated risk of a multitude of cancers and increased rates of psychopathology, even at very low levels of arsenic exposure. The hypothalamic-pituitary-adrenal (HPA) axis represents a multisite integration center that regulates a wide scope of biological and physiological processes: breakdown within this system can generate an array of far-reaching effects, making it an intriguing candidate for arsenic-mediated damage. Using a mouse model, we examined the effects of perinatal exposure to 50 ppb sodium arsenate on the functioning of the HPA axis through the assessment of corticotrophin-releasing factor (CRF), proopiomelanocortin (Pomc) mRNA, adrenocorticotrophin hormone (ACTH), corticosterone (CORT), 11ß-hydroxysteroid dehydrogenase Type 1 (11ß-HSD 1), and glucocorticoid receptor (GR) protein and mRNA. Compared to controls, we observed that the perinatal arsenic-exposed offspring exhibit an increase in hypothalamic CRF, altered CORT secretion both at baseline and in response to a stressor, decreased hippocampal 11ß-HSD 1 and altered subcellular GR distribution in the hypothalamus. These data indicate significant HPA axis impairment at post-natal day 35 resulting from perinatal exposure to 50 ppb sodium arsenate. Our findings suggest that the dysregulation of this critical regulatory axis could underlie important molecular and cognitive pathology observed following exposure to arsenic.


Assuntos
Arseniatos/toxicidade , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Poluentes da Água/toxicidade , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Hormônio Adrenocorticotrópico/sangue , Hormônio Adrenocorticotrópico/genética , Análise de Variância , Animais , Animais Recém-Nascidos , Ensaio de Imunoadsorção Enzimática , Comportamento Alimentar/efeitos dos fármacos , Hipoxantina Fosforribosiltransferase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/metabolismo
6.
Neurotoxicol Teratol ; 33(5): 530-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21784148

RESUMO

Changes within the glucocorticoid receptor (GR) cellular signaling pathway were evaluated in adolescent mice exposed to 50 ppb arsenic during gestation. Previously, we reported increased basal plasma corticosterone levels, decreased hippocampal GR levels and deficits in learning and memory performance in perinatal arsenic-exposed mice. The biosynthesis of members of the mitogen-activated protein kinase (MAPK) signaling pathway, known to be involved in learning and memory, is modulated by the binding of GR to glucocorticoid response elements (GREs) in the gene promoters. Two genes of the MAPK pathway, Ras and Raf, contain GREs which are activated upon binding of GRs. We evaluated the activity of GRs at Ras and Raf promoters using chromatin immunoprecipitation and real-time PCR and report decreased binding of the GR at these promoters. An ELISA-based GR binding assay was used to explore whether this decreased binding was restricted to in vivo promoters and revealed no differences in binding of native GR to synthetic GREs. The decreased in vivo GR binding coincides with significantly decreased mRNA levels and slight reductions of protein of both H-Ras and Raf-1 in perinatally arsenic-exposed mice. Nuclear activated extracellular-signal regulated kinase (ERK), a downstream target of Ras and Raf, whose transcriptional targets also play an important role in learning and memory, was decreased in the hippocampus of arsenic-exposed animals when compared to controls. GR-mediated transcriptional deficits in the MAPK/ERK pathway could be an underlying cause of previously reported learning deficits and provide the link to arsenic-induced deficiencies in cognitive development.


Assuntos
Arsênio/toxicidade , Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta/efeitos dos fármacos , Animais , Western Blotting/métodos , Imunoprecipitação da Cromatina/métodos , MAP Quinases Reguladas por Sinal Extracelular/biossíntese , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Proteínas Proto-Oncogênicas c-raf/biossíntese , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Elementos de Resposta/genética , Transdução de Sinais/genética
7.
Nat Med ; 17(5): 559-65, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21516088

RESUMO

Deficiency in fragile X mental retardation protein (FMRP) results in fragile X syndrome (FXS), an inherited form of intellectual disability. Despite extensive research, it is unclear how FMRP deficiency contributes to the cognitive deficits in FXS. Fmrp-null mice show reduced adult hippocampal neurogenesis. As Fmrp is also enriched in mature neurons, we investigated the function of Fmrp expression in neural stem and progenitor cells (aNSCs) and its role in adult neurogenesis. Here we show that ablation of Fmrp in aNSCs by inducible gene recombination leads to reduced hippocampal neurogenesis in vitro and in vivo, as well as markedly impairing hippocampus-dependent learning in mice. Conversely, restoration of Fmrp expression specifically in aNSCs rescues these learning deficits in Fmrp-deficient mice. These data suggest that defective adult neurogenesis may contribute to the learning impairment seen in FXS, and these learning deficits can be rectified by delayed restoration of Fmrp specifically in aNSCs.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/fisiologia , Síndrome do Cromossomo X Frágil/fisiopatologia , Hipocampo/fisiopatologia , Aprendizagem/fisiologia , Células-Tronco Adultas/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/patologia , Síndrome do Cromossomo X Frágil/psicologia , Hipocampo/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neurogênese/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...