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










Base de datos
Intervalo de año de publicación
1.
Neurotoxicol Teratol ; 59: 1-15, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27751817

RESUMEN

Exposure to arsenic, a common environmental toxin found in drinking water, leads to a host of neurological pathologies. We have previously demonstrated that developmental exposure to a low level of arsenic (50ppb) alters epigenetic processes that underlie deficits in adult hippocampal neurogenesis leading to aberrant behavior. It is unclear if arsenic impacts the programming and regulation of embryonic neurogenesis during development when exposure occurs. The master negative regulator of neural-lineage, REST/NRSF, controls the precise timing of fate specification and differentiation of neural stem cells (NSCs). Early in development (embryonic day 14), we observed increased expression of Rest, its co-repressor, CoREST, and the inhibitory RNA binding/splicing protein, Ptbp1, and altered expression of mRNA spliced isoforms of Pbx1 that are directly regulated by these factors in the male brain in response to prenatal 50ppb arsenic exposure. These increases were concurrent with decreased expression of microRNA-9 (miR-9), miR-9*, and miR-124, all of which are REST/NRSF targets and inversely regulate Rest expression to allow for maturation of NSCs. Exposure to arsenic decreased the formation of neuroblasts in vitro from NSCs derived from male pup brains. The female response to arsenic was limited to increased expression of CoREST and Ptbp2, an RNA binding protein that allows for appropriate splicing of genes involved in the progression of neurogenesis. These changes were accompanied by increased neuroblast formation in vitro from NSCs derived from female pups. Unexposed male mice express transcriptomic factors to induce differentiation earlier in development compared to unexposed females. Thus, arsenic exposure likely delays differentiation of NSCs in males while potentially inducing precocious differentiation in females early in development. These effects are mitigated by embryonic day 18 of development. Arsenic-induced dysregulation of the regulatory loop formed by REST/NRSF, its target microRNAs, miR-9 and miR-124, and RNA splicing proteins, PTBP1 and 2, leads to aberrant programming of NSC function that is perhaps perpetuated into adulthood inducing deficits in differentiation we have previously observed.


Asunto(s)
Arsénico/toxicidad , Ribonucleoproteínas Nucleares Heterogéneas/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Células-Madre Neurales/efectos de los fármacos , Proteína de Unión al Tracto de Polipirimidina/biosíntesis , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteínas Represoras/biosíntesis , Caracteres Sexuales , Animales , Células Cultivadas , Proteínas Co-Represoras , Femenino , Masculino , Ratones , MicroARNs/biosíntesis , Neurogénesis/efectos de los fármacos , Factor de Transcripción 1 de la Leucemia de Células Pre-B/biosíntesis , Embarazo
2.
Alcohol Clin Exp Res ; 39(4): 631-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25752869

RESUMEN

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.


Asunto(s)
Etanol/efectos adversos , Hipocampo/efectos de los fármacos , Vivienda para Animales , Efectos Tardíos de la Exposición Prenatal/psicología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Discriminación en Psicología/efectos de los fármacos , Femenino , Hipocampo/metabolismo , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo
3.
Toxicol Rep ; 2: 1376-1390, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26855884

RESUMEN

Previously we have shown that prenatal moderate arsenic exposure (50 ppb) disrupts glucocorticoid receptor (GR) programming and that these changes continue into adolescence in males. However, it was not clear what the molecular mechanisms were promoting these GR programming changes or if these changes occurred in arsenic-exposed females. In the present studies, we assessed the effects of arsenic on protein and mRNA of the glucocorticoid receptor (GR) and 11ß-hydroxysteroid dehydrogenase (Hsd) isozymes and compared the levels of methylation within the promoters of the Nr3c1 and Hsd11b1 genes in female fetal brain at embryonic days (E) 14 and 18. Prenatal arsenate exposure produced sex specific effects on the glucocorticoid system. Compared to males, females were resistant to arsenic induced changes in GR, 11ß-Hsd-1 and 11ß-Hsd-2 protein levels despite observed elevations in Nr3c1 and Hsd11b2 mRNA. This sex-specific effect was not due to differences in the methylation of the GR promoter as methylation of the Nr3c1 gene was either unchanged (region containing the egr-1 binding site) or similarly reduced (region containing the SP-1 transcription factor binding site) in both males and females exposed to arsenic. Arsenic did produce sex and age-specific changes in the methylation of Hsd11b1 gene, producing increased methylation in females at E14 and decreased methylation at E18. These changes were not attributed to changes in DNMT levels. Since arsenate metabolism could interfere with the generation of methyl donor groups, we assessed glutathione (GSH), s-adenosylmethionine (SAM) and As 3 methyltransferase (As3MT). Exposed males and females had similar levels of As3MT and SAM; however, females had higher levels of GSH/GSSH. It is possible that this greater anti-oxidative capacity within the females provides protection against low to moderate arsenate. Our data suggest that the GR signaling system in female offspring was not as affected by prenatal arsenic and predicts that female arsenic-exposed mice should have normal GR feedback regulation.

4.
Artículo en Inglés | MEDLINE | ID: mdl-26989786

RESUMEN

Research identifying connections between the gastrointestinal flora and human health has developed at a rapid pace. Several studies link the gut microbiome to a variety of biological functions beyond the gastrointestinal tract. Changes in our diets, including the consumption of artificial sweeteners, have profound effects on the composition of the gut microbiome and can, in turn, affect brain function, glucose tolerance, and inflammation. Sweeteners are often used to encourage consumption of agents such as ethanol and nicotine in laboratory studies using rodents. Studies aiming to examine the effects of agents like ethanol on the developing nervous system administer these agents during pregnancy. To date, there have been no studies exploring the impact of the combination of dietary ethanol and saccharin during pregnancy on the gut microbiome in either humans or laboratory animal models. In the study presented, we evaluated the impact of ethanol in either water or saccharin on the fecal microbiome in pregnant and non-pregnant mice using a qPCR approach. We found that the combination of ethanol and saccharin produced different effects than ethanol in water, depending on pregnancy status. Levels of Clostridium were reduced in ethanol-saccharin but not ethanol-water drinking mice, even though the total levels of ethanol consumed were the same for the two groups. Eubacteria were increased in the pregnant, but decreased in the non-pregnant, ethanol-saccharin drinking group. These treatment and pregnancy specific changes could impact the development of the offspring. In developing and quality checking our primer sets for these studies we identified several problems within previous research in the field. The technical drawbacks in previous studies, as well as our own study, are discussed. Despite some progress in the ability to study the gut microbiome, more advances and standardization of practices should be established to improve the reliability and validity of microbiome research.

5.
Neurotoxicol Teratol ; 47: 66-79, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25459689

RESUMEN

The glucocorticoid system, which plays a critical role in a host of cellular functions including mood disorders and learning and memory, has been reported to be disrupted by arsenic. In previous work we have developed and characterized a prenatal moderate arsenic exposure (50ppb) model and identified several deficits in learning and memory and mood disorders, as well as alterations within the glucocorticoid receptor signaling system in the adolescent mouse. In these present studies we assessed the effects of arsenic on the glucocorticoid receptor (GR) pathway in both the placenta and the fetal brain in response at two critical periods, embryonic days 14 and 18. The focus of these studies was on the 11ß-hydroxysteroid dehydrogenase enzymes (11ß-HSD1 and 11ß-HSD2) which play a key role in glucorticoid synthesis, as well as the expression and set point of the GR negative feedback regulation. Negative feedback regulation is established early in development. At E14 we found arsenic exposure significantly decreased expression of both protein and message in brain of GR and the 11ß-HSD1, while 11ß-HSD2 enzyme protein levels were increased but mRNA levels were decreased in the brain. These changes in brain protein continued into the E18 time point, but mRNA levels were no longer significantly altered. Placental HSD11B2 mRNA was not altered by arsenic treatment but protein levels were elevated at E14. GR placental protein levels were decreased at E18 in the arsenic exposed condition. This suggests that arsenic exposure may alter GR expression levels as a consequence of a prolonged developmental imbalance between 11ß-HSD1 and 11ß-HSD2 protein expression despite decreased 11HSDB2 mRNA. The suppression of GR and the failure to turn down 11ß-HSD2 protein expression during fetal development may lead to an altered set point for GR signaling throughout adulthood. To our knowledge, these studies are the first to demonstrate that gestational exposure to moderate levels of arsenic results in altered fetal programming of the glucocorticoid system.


Asunto(s)
Arsénico/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Glucocorticoides/sangre , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Transducción de Señal/efectos de los fármacos , Factores de Edad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Placenta/efectos de los fármacos , Placenta/embriología , Placenta/patología , Embarazo , ARN Mensajero/metabolismo , Caracteres Sexuales , Proteínas de Unión a Tacrolimus/metabolismo
6.
Neurotoxicology ; 33(5): 1338-45, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22960421

RESUMEN

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.


Asunto(s)
Arseniatos/toxicidad , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Contaminantes del Agua/toxicidad , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Ensayo de Inmunoadsorción Enzimática , Conducta Alimentaria/efectos de los fármacos , Hipoxantina Fosforribosiltransferasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/metabolismo
7.
Neurotoxicol Teratol ; 33(5): 530-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21784148

RESUMEN

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.


Asunto(s)
Arsénico/toxicidad , Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/efectos de los fármacos , Animales , Western Blotting/métodos , Inmunoprecipitación de Cromatina/métodos , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Elementos de Respuesta/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA