Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Environ Epigenet ; 4(2): dvy002, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29732168

RESUMEN

Early life exposures to endocrine-disrupting chemicals (EDCs) have been associated with physiological changes of endocrine-sensitive tissues throughout postnatal life. Although hormones play a critical role in skeletal growth and maintenance, the effects of prenatal EDC exposure on adult bone health are not well understood. Moreover, studies assessing skeletal changes across multiple generations are limited. In this article, we present previously unpublished data demonstrating dose-, sex-, and generation-specific changes in bone morphology and function in adult mice developmentally exposed to the model estrogenic EDC bisphenol A (BPA) at doses of 10 µg (lower dose) or 10 mg per kg bw/d (upper dose) throughout gestation and lactation. We show that F1 generation adult males, but not females, developmentally exposed to bisphenol A exhibit dose-dependent reductions in outer bone size resulting in compromised bone stiffness and strength. These structural alterations and weaker bone phenotypes in the F1 generation did not persist in the F2 generation. Instead, F2 generation males exhibited greater bone strength. The underlying mechanisms driving the EDC-induced physiological changes remain to be determined. We discuss potential molecular changes that could contribute to the EDC-induced skeletal effects, with an emphasis on epigenetic dysregulation. Furthermore, we assess the necessity of intact sex steroid receptors to mediate these effects. Expanding future assessments of EDC-induced effects to the skeleton may provide much needed insight into one of the many health effects of these chemicals and aid in regulatory decision making regarding exposure of vulnerable populations to these chemicals.

2.
Horm Behav ; 102: 93-104, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29763587

RESUMEN

Fetal exposure to endocrine disrupting chemicals (EDCs) has been associated with adverse neurobehavioral outcomes across the lifespan and can persist across multiple generations of offspring. However, the underlying mechanisms driving these changes are not well understood. We investigated the molecular perturbations associated with EDC-induced behavioral changes in first (F1) and second (F2) filial generations, using the model EDC bisphenol A (BPA). C57BL/6J dams were exposed to BPA from preconception until lactation through the diet at doses (10 µg/kg bw/d-lower dose or 10 mg/kg bw/d-upper dose) representative of human exposure levels. As adults, F1 male offspring exhibited increased depressive-like behavior, measured by the forced swim test, while females were unaffected. These behavioral changes were limited to the F1 generation and were not associated with altered maternal care. Transcriptome analysis by RNA-sequencing in F1 control and upper dose BPA-exposed adult male hippocampus revealed neurotransmitter systems as major pathways disrupted by developmental BPA exposure. High performance liquid chromatography demonstrated a male-specific reduction in hippocampal serotonin. Administration of the selective serotonin reuptake inhibitor fluoxetine (20 mg/kg bw) rescued the depressive-like phenotype in males exposed to lower, but not upper, dose BPA, suggesting distinct mechanisms of action for each exposure dose. Finally, high resolution mass spectrometry revealed reduced circulating levels of the neuroactive steroid dehydroepiandrosterone in BPA-exposed males, suggesting another potential mechanism underlying the depressive-like phenotype. Thus, behavioral changes associated with early life BPA exposure may be mediated by sex-specific disruptions in the serotonergic system and/or sex steroid biogenesis in male offspring.


Asunto(s)
Conducta Animal/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Depresión/inducido químicamente , Disruptores Endocrinos/farmacología , Hipotálamo/efectos de los fármacos , Conducta Materna/efectos de los fármacos , Fenoles/farmacología , Efectos Tardíos de la Exposición Prenatal/psicología , Animales , Fármacos del Sistema Nervioso Central/metabolismo , Depresión/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neurotransmisores/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Esteroides/metabolismo
3.
Environ Health Perspect ; 125(9): 097022, 2017 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-29161229

RESUMEN

BACKGROUND: Exposure to the environmental endocrine disruptor bisphenol A (BPA) is ubiquitous and associated with the increased risk of diabetes and obesity. However, the underlying mechanisms remain unknown. We recently demonstrated that perinatal BPA exposure is associated with higher body fat, impaired glucose tolerance, and reduced insulin secretion in first- (F1) and second-generation (F2) C57BL/6J male mice offspring. OBJECTIVE: We sought to determine the multigenerational effects of maternal bisphenol A exposure on mouse pancreatic islets. METHODS: Cellular and molecular mechanisms underlying these persistent changes were determined in F1 and F2 adult offspring of F0 mothers exposed to two relevant human exposure levels of BPA (10µg/kg/d-LowerB and 10mg/kg/d-UpperB). RESULTS: Both doses of BPA significantly impaired insulin secretion in male but not female F1 and F2 offspring. Surprisingly, LowerB and UpperB induced islet inflammation in male F1 offspring that persisted into the next generation. We also observed dose-specific effects of BPA on islets in males. UpperB exposure impaired mitochondrial function, whereas LowerB exposure significantly reduced ß-cell mass and increased ß-cell death that persisted in the F2 generation. Transcriptome analyses supported these physiologic findings and there were significant dose-specific changes in the expression of genes regulating inflammation and mitochondrial function. Previously we observed increased expression of the critically important ß-cell gene, Igf2 in whole F1 embryos. Surprisingly, increased Igf2 expression persisted in the islets of male F1 and F2 offspring and was associated with altered DNA methylation. CONCLUSION: These findings demonstrate that maternal BPA exposure has dose- and sex-specific effects on pancreatic islets of adult F1 and F2 mice offspring. The transmission of these changes across multiple generations may involve either mitochondrial dysfunction and/or epigenetic modifications. https://doi.org/10.1289/EHP1674.


Asunto(s)
Compuestos de Bencidrilo/efectos adversos , Disruptores Endocrinos/efectos adversos , Contaminantes Ambientales/efectos adversos , Islotes Pancreáticos/efectos de los fármacos , Exposición Materna/efectos adversos , Fenoles/efectos adversos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Relación Dosis-Respuesta a Droga , Epigénesis Genética , Femenino , Insulina/metabolismo , Secreción de Insulina , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Factores Sexuales
4.
Endocrinology ; 158(8): 2533-2542, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28549143

RESUMEN

Increasing evidence has demonstrated that exposure to endocrine-disrupting chemicals impacts maternal and fetal health, but the underlying mechanisms are still unclear. We previously showed that dietary exposure to 10 µg/kg body weight (bw)/d and 10 mg/kg bw/d of bisphenol A (BPA) during pregnancy induced metabolic abnormalities in F1 male offspring and gestational glucose intolerance in F0 pregnant mice. The aim of this study was to elucidate the underlying etiologies of BPA exposure-induced metabolic disease by analyzing the male fetal liver metabolome. Using the Metabolon Discover HD4 Platform, our laboratory identified metabolic pathways that were altered by BPA exposure, including biochemicals in lipid and amino acid metabolism. Specifically, primary and secondary bile acids were increased in liver from BPA-exposed embryonic day 18.5 male fetuses. We subsequently showed that increased bile acid was associated with a defective farnesoid X receptor-dependent negative feedback mechanism in BPA-exposed fetuses. In addition, through metabolomics, we observed that BPA-exposed fetuses had elevated tryptophan levels. Independent liquid chromatography and mass spectrometry measurement revealed that BPA-exposed dams also had increased tryptophan levels relative to those of controls. Because several key enzymes in tryptophan catabolism are vitamin B6 dependent and vitamin B6 deficiencies have been linked to gestational diabetes, we tested the impact of vitamin B6 supplementation and showed that it rescued gestational glucose intolerance in BPA-exposed pregnant mice. Our study has therefore identified two pathways (bile acid and tryptophan metabolism) that potentially underlie BPA-induced maternal and fetal metabolic disease.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Ácidos y Sales Biliares/metabolismo , Estrógenos no Esteroides/toxicidad , Fenoles/toxicidad , Triptófano/metabolismo , Animales , Compuestos de Bencidrilo/administración & dosificación , Relación Dosis-Respuesta a Droga , Estrógenos no Esteroides/administración & dosificación , Femenino , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Fenoles/administración & dosificación , Embarazo , Efectos Tardíos de la Exposición Prenatal
5.
Fertil Steril ; 106(4): 930-40, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27523298

RESUMEN

In mammals, the extraembryonic tissues, which include the placenta, are crucial for embryonic development and growth. Because the placenta is no longer needed for postnatal life, however, it has been relatively understudied as a tissue of interest in biomedical research. Recently, increased efforts have been placed on understanding the placenta and how it may play a key role in human health and disease. In this review, we discuss two very different types of environmental exposures: assisted reproductive technologies and in utero exposure to endocrine-disrupting chemicals. We summarize the current literature on their effects on placental development in both rodent and human, and comment on the potential use of placental biomarkers as predictors of offspring health outcomes.


Asunto(s)
Disruptores Endocrinos/efectos adversos , Contaminantes Ambientales/efectos adversos , Fármacos para la Fertilidad Femenina/efectos adversos , Exposición Materna/efectos adversos , Placenta/efectos de los fármacos , Reproducción/efectos de los fármacos , Técnicas Reproductivas Asistidas/efectos adversos , Animales , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Impresión Genómica/efectos de los fármacos , Humanos , Placenta/metabolismo , Placenta/patología , Embarazo , Medición de Riesgo , Factores de Riesgo
6.
Semin Cell Dev Biol ; 43: 66-75, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26026600

RESUMEN

Increasing evidence has highlighted the critical role of early life environment in shaping the future health outcomes of an individual. Moreover, recent studies have revealed that early life perturbations can affect the health of subsequent generations. Hypothesized mechanisms of multi- and transgenerational inheritance of abnormal developmental phenotypes include epigenetic misregulation in germ cells. In this review, we will focus on the available data demonstrating the ability of endocrine disrupting chemicals (EDCs), including bisphenol A (BPA), phthalates, and parabens, to alter epigenetic marks in rodents and humans. These epigenetic marks include DNA methylation, histone post-translational modifications, and non-coding RNAs. We also review the current evidence for multi- and transgenerational inheritance of abnormal developmental changes in the offspring following EDC exposure. Based on published results, we conclude that EDC exposure can alter the mouse and human epigenome, with variable tissue susceptibilities. Although increasing data suggest that exposure to EDCs is linked to transgenerational inheritance of reproductive, metabolic, or neurological phenotypes, more studies are needed to validate these observations and to elucidate further whether these developmental changes are directly associated with the relevant epigenetic alterations.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Disruptores Endocrinos/farmacología , Epigénesis Genética/efectos de los fármacos , Patrón de Herencia/efectos de los fármacos , Parabenos/farmacología , Fenoles/farmacología , Ácidos Ftálicos/farmacología , Animales , Compuestos de Bencidrilo/efectos adversos , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Disruptores Endocrinos/efectos adversos , Epigénesis Genética/genética , Humanos , Ratones , Parabenos/efectos adversos , Fenoles/efectos adversos , Fenotipo , Ácidos Ftálicos/efectos adversos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , ARN no Traducido/efectos de los fármacos , ARN no Traducido/genética
7.
Endocrinology ; 156(6): 2049-58, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25807043

RESUMEN

Accumulating evidence has suggested that a suboptimal early life environment produces multigenerational developmental defects. A proposed mechanism is stable inheritance of DNA methylation. Here we show that maternal bisphenol A (BPA) exposure in C57BL/6 mice produces multigenerational metabolic phenotypes in their offspring. Using various methods including dual-energy X-ray absorptiometry analyses, glucose tolerance tests, and perifusion islet studies, we showed that exposure to 10 µg/kg/d and 10 mg/kg/d BPA in pregnant F0 mice was associated with higher body fat and perturbed glucose homeostasis in F1 and F2 male offspring but not female offspring. To provide insight into the mechanism of the multigenerational metabolic abnormalities, we investigated the maternal metabolic milieu and inheritance of DNA methylation across generations. We showed that maternal glucose homeostasis during pregnancy was altered in the F0 but not F1 female mice. The results suggested that a compromised maternal metabolic milieu may play a role in the health of the F1 offspring but cannot account for all of the observed multigenerational phenotypes. We further demonstrated that the metabolic phenotypes in the F1 and F2 BPA male offspring were linked to fetal overexpression of the imprinted Igf2 gene and increased DNA methylation at the Igf2 differentially methylated region 1. Studies in H19(Δ3.8/+) mouse mutants supported the role of fetal Igf2 overexpression in altered adult glucose homeostasis. We conclude that early life BPA exposure at representative human exposure levels can perturb metabolic health across multiple generations in the mouse through stable inheritance of DNA methylation changes at the Igf2 locus.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Fenoles/toxicidad , Animales , Peso al Nacer/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Femenino , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología
8.
Mol Ther ; 22(6): 1084-1095, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24662946

RESUMEN

Rhesus (rh) but not human (hu) TRIM5α potently restricts human immunodeficiency virus (HIV)-1 infection. It is not clear why huTRIM5α fails to effectively block HIV infection, but it is thought to have a lower affinity for the viral core. Using primary human CD4 T cells, we investigated the ability of huTRIM5α, rhTRIM5α, and the huTRIM5αR323-332 B30.2/SPRY patch-mutant to form cytoplasmic bodies, postulated as key components of the HIV-1 restriction apparatus. Both rhTRIM5α and huTRIM5αR323-332 formed pronounced cytoplasmic bodies, whereas cytoplasmic bodies in T cells overexpressing huTRIM5α were present but more difficult to detect. As expression of all three TRIM5α orthologs was similar at the RNA level, we next investigated the role of protein stability in conferring TRIM5α-mediated HIV-1 restriction. Both steady-state and pulse-chase experiments revealed that the huTRIM5α protein was much less stable than rhTRIM5α, and this difference correlated with higher self-ubiquitination activity. Using a stabilized form of huTRIM5α in which the steady-state expression level was more similar to rhTRIM5α, we observed comparable HIV-1 restriction activity in multi-round HIV-1 challenge assays. Lastly, primary human CD4 T cells expressing a stabilized huTRIM5α were protected from HIV-1-mediated destruction in vivo, indicating that efforts to stabilize huTRIM5α should have significant long-term therapeutic value.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Infecciones por VIH/prevención & control , VIH-1/inmunología , Proteínas/genética , Proteínas/metabolismo , Animales , Factores de Restricción Antivirales , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Infecciones por VIH/metabolismo , VIH-1/crecimiento & desarrollo , Humanos , Macaca mulatta , Ratones , Estabilidad Proteica , ARN/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Ubiquitinación
9.
Cell Cycle ; 13(2): 199-207, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24200970

RESUMEN

Epithelial ovarian cancer (EOC) is the leading cause of gynecological-related cancer deaths in the United States. There is, therefore, an urgent need to develop novel therapeutic strategies for this devastating disease. Cellular senescence is a state of stable cell growth arrest that acts as an important tumor suppression mechanism. Ribonucleotide reductase M2 (RRM2) plays a key role in regulating the senescence-associated cell growth arrest by controlling biogenesis of 2'-deoxyribonucleoside 5'-triphosphates (dNTPs). The role of RRM2 in EOC remains poorly understood. Here we show that RRM2 is expressed at higher levels in EOCs compared with either normal ovarian surface epithelium (P<0.001) or fallopian tube epithelium (P<0.001). RRM2 expression significantly correlates with the expression of Ki67, a marker of cell proliferation (P<0.001). Moreover, RRM2 expression positively correlates with tumor grade and stage, and high RRM2 expression independently predicts a shorter overall survival in EOC patients (P<0.001). To delineate the functional role of RRM2 in EOC, we knocked down RRM2 expression in a panel of EOC cell lines. Knockdown of RRM2 expression inhibits the growth of human EOC cells. Mechanistically, RRM2 knockdown triggers cellular senescence in these cells. Notably, this correlates with the induction of the DNA damage response, a known mediator of cellular senescence. These data suggest that targeting RRM2 in EOCs by suppressing its activity is a novel pro-senescence therapeutic strategy that has the potential to improve survival of EOC patients.


Asunto(s)
Senescencia Celular , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Ováricas/enzimología , Ribonucleósido Difosfato Reductasa/metabolismo , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , Epitelio/enzimología , Trompas Uterinas/enzimología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Antígeno Ki-67/metabolismo , Persona de Mediana Edad , Terapia Molecular Dirigida , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Ovario/enzimología , Ribonucleósido Difosfato Reductasa/genética , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA