Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Pharmacol Toxicol ; 52: 455-79, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22017681

RESUMEN

Puberty is an important transition that enables reproduction of mammalian species. Precocious puberty, specifically early thelarche (the appearance of breast "buds"), in girls of multiple ethnic backgrounds is a major health problem in the United States and other countries. The cause for a continued decrease in the age of breast development in girls is unknown, but environmental factors likely play a major role. Laboratory and epidemiological studies have identified several individual environmental factors that affect breast development, but further progress is needed. Current research needs include increased attention to and recording of prenatal and neonatal environmental exposures, testing of marketed chemicals for effects on the mammary gland, and understanding of the mammary gland-specific mechanisms that are altered by chemicals. Such research is required to halt the increasing trend toward puberty at earlier ages.


Asunto(s)
Neoplasias de la Mama/inducido químicamente , Mama/efectos de los fármacos , Exposición a Riesgos Ambientales/análisis , Animales , Peso Corporal , Mama/patología , Dieta , Disruptores Endocrinos/toxicidad , Contaminantes Ambientales/toxicidad , Femenino , Humanos , Atención Perinatal , Pubertad/efectos de los fármacos , Pubertad/fisiología , Pubertad Precoz/inducido químicamente , Pubertad Precoz/patología , Reproducción/efectos de los fármacos , Factores de Riesgo , Roedores/crecimiento & desarrollo , Estados Unidos
2.
Environ Health ; 13(1): 25, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24690217

RESUMEN

BACKGROUND: Human exposure to bisphenol A (BPA) is ubiquitous, yet there are concerns about whether BPA can be measured in human blood. This Round Robin was designed to address this concern through three goals: 1) to identify collection materials, reagents and detection apparatuses that do not contribute BPA to serum; 2) to identify sensitive and precise methods to accurately measure unconjugated BPA (uBPA) and BPA-glucuronide (BPA-G), a metabolite, in serum; and 3) to evaluate whether inadvertent hydrolysis of BPA-G occurs during sample handling and processing. METHODS: Four laboratories participated in this Round Robin. Laboratories screened materials to identify BPA contamination in collection and analysis materials. Serum was spiked with concentrations of uBPA and/or BPA-G ranging from 0.09-19.5 (uBPA) and 0.5-32 (BPA-G) ng/mL. Additional samples were preserved unspiked as 'environmental' samples. Blinded samples were provided to laboratories that used LC/MSMS to simultaneously quantify uBPA and BPA-G. To determine whether inadvertent hydrolysis of BPA metabolites occurred, samples spiked with only BPA-G were analyzed for the presence of uBPA. Finally, three laboratories compared direct and indirect methods of quantifying BPA-G. RESULTS: We identified collection materials and reagents that did not introduce BPA contamination. In the blinded spiked sample analysis, all laboratories were able to distinguish low from high values of uBPA and BPA-G, for the whole spiked sample range and for those samples spiked with the three lowest concentrations (0.5-3.1 ng/ml). By completion of the Round Robin, three laboratories had verified methods for the analysis of uBPA and two verified for the analysis of BPA-G (verification determined by: 4 of 5 samples within 20% of spiked concentrations). In the analysis of BPA-G only spiked samples, all laboratories reported BPA-G was the majority of BPA detected (92.2 - 100%). Finally, laboratories were more likely to be verified using direct methods than indirect ones using enzymatic hydrolysis. CONCLUSIONS: Sensitive and accurate methods for the direct quantification of uBPA and BPA-G were developed in multiple laboratories and can be used for the analysis of human serum samples. BPA contamination can be controlled during sample collection and inadvertent hydrolysis of BPA conjugates can be avoided during sample handling.


Asunto(s)
Compuestos de Bencidrilo/sangre , Contaminantes Ambientales/sangre , Fenoles/sangre , Animales , Recolección de Muestras de Sangre/métodos , Cromatografía Líquida de Alta Presión , Monitoreo del Ambiente , Glucurónidos/sangre , Humanos , Laboratorios , Ratas , Espectrometría de Masas en Tándem
3.
Environ Health Perspect ; 132(4): 45001, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38592230

RESUMEN

BACKGROUND: The European Food Safety Authority (EFSA) recommended lowering their estimated tolerable daily intake (TDI) for bisphenol A (BPA) 20,000-fold to 0.2 ng/kg body weight (BW)/day. BPA is an extensively studied high production volume endocrine disrupting chemical (EDC) associated with a vast array of diseases. Prior risk assessments of BPA by EFSA as well as the US Food and Drug Administration (FDA) have relied on industry-funded studies conducted under good laboratory practice protocols (GLP) requiring guideline end points and detailed record keeping, while also claiming to examine (but rejecting) thousands of published findings by academic scientists. Guideline protocols initially formalized in the mid-twentieth century are still used by many regulatory agencies. EFSA used a 21st century approach in its reassessment of BPA and conducted a transparent, but time-limited, systematic review that included both guideline and academic research. The German Federal Institute for Risk Assessment (BfR) opposed EFSA's revision of the TDI for BPA. OBJECTIVES: We identify the flaws in the assumptions that the German BfR, as well as the FDA, have used to justify maintaining the TDI for BPA at levels above what a vast amount of academic research shows to cause harm. We argue that regulatory agencies need to incorporate 21st century science into chemical hazard identifications using the CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity) nonguideline academic studies in a collaborative government-academic program model. DISCUSSION: We strongly endorse EFSA's revised TDI for BPA and support the European Commission's (EC) apparent acceptance of this updated BPA risk assessment. We discuss challenges to current chemical risk assessment assumptions about EDCs that need to be addressed by regulatory agencies to, in our opinion, become truly protective of public health. Addressing these challenges will hopefully result in BPA, and eventually other structurally similar bisphenols (called regrettable substitutions) for which there are known adverse effects, being eliminated from all food-related and many other uses in the EU and elsewhere. https://doi.org/10.1289/EHP13812.


Asunto(s)
Compuestos de Bencidrilo , Fenoles , Humanos , Inocuidad de los Alimentos , Nivel sin Efectos Adversos Observados , Revisiones Sistemáticas como Asunto
4.
Birth Defects Res ; 115(15): 1345-1397, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37646438

RESUMEN

BACKGROUND: The Consortium Linking Academic and Regulatory Insights on Bisphenol A Toxicity (CLARITY-BPA) was a collaborative research effort to better link academic research with governmental guideline studies. This review explores the secondary goal of CLARITY-BPA: to identify endpoints or technologies from CLARITY-BPA and prior/concurrent literature from these laboratories that may enhance the capacity of rodent toxicity studies to detect endocrine disrupting chemicals (EDCs). METHODS: A systematic literature search was conducted with search terms for BPA and the CLARITY-BPA participants. Relevant studies employed a laboratory rodent model and reported results on 1 of the 10 organs/organ systems evaluated in CLARITY-BPA (brain and behavior, cardiac, immune, mammary gland, ovary, penile function, prostate gland and urethra, testis and epididymis, thyroid hormone and metabolism, and uterus). Study design and findings were summarized, and a risk-of-bias assessment was conducted. RESULTS: Several endpoints and methods were identified as potentially helpful to detect effects of EDCs. For example, molecular and quantitative morphological approaches were sensitive in detecting alterations in early postnatal development of the brain, ovary, and mammary glands. Hormone challenge studies mimicking human aging reported increased susceptibility of the prostate to disease following developmental BPA exposure. Statistical analyses for nonmonotonic dose responses, and computational approaches assessing multiple treatment-related outcomes concurrently in linked hormone-sensitive organ systems, reported effects at low BPA doses. CONCLUSIONS: This review provided an opportunity to evaluate the unique insights provided by nontraditional assessments in CLARITY-BPA to identify technologies and endpoints to enhance detection of EDCs in future studies.


Asunto(s)
Disruptores Endocrinos , Masculino , Femenino , Humanos , Disruptores Endocrinos/toxicidad , Organizaciones , Compuestos de Bencidrilo/toxicidad , Fenoles/toxicidad
7.
Hormones (Athens) ; 9(3): 206-17, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20688618

RESUMEN

Environmental chemicals with hormone-like activity can disrupt programming of endocrine signaling pathways during development and result in adverse effects, some of which may not be apparent until much later in life. Recent reports link exposure to environmental endocrine disrupting chemicals during development with adverse health consequences, including obesity and diabetes. These particular diseases are quickly becoming significant public health problems and are fast reaching epidemic proportions worldwide. This review summarizes data from experimental animals and humans which support an association of endocrine disrupting chemicals, such as diethylstilbestrol, bisphenol A, phytoestrogens, phthalates, and organotins, with the development of obesity. Potential mechanisms are summarized and future research needs are discussed.


Asunto(s)
Disruptores Endocrinos/efectos adversos , Contaminantes Ambientales/efectos adversos , Obesidad/inducido químicamente , Animales , Humanos , Obesidad/fisiopatología , Medición de Riesgo , Factores de Riesgo , Aumento de Peso/efectos de los fármacos
8.
Biol Reprod ; 81(4): 690-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19535786

RESUMEN

Developmental exposure to endocrine-disrupting compounds is hypothesized to adversely affect female reproductive physiology by interfering with the organization of the hypothalamic-pituitary-gonadal axis. Here, we compared the effects of neonatal exposure to two environmentally relevant doses of the plastics component bisphenol-A (BPA; 50 microg/kg and 50 mg/kg) with the ESR1 (formerly known as ERalpha)-selective agonist 4,4',4''-(4-propyl-[(1)H]pyrazole-1,3,5-triyl)trisphenol (PPT; 1 mg/kg) on the development of the female rat hypothalamus and ovary. An oil vehicle and estradiol benzoate (EB; 25 microg) were used as negative and positive controls. Exposure to EB, PPT, or the low dose of BPA advanced pubertal onset. A total of 67% of females exposed to the high BPA dose were acyclic by 15 wk after vaginal opening compared with 14% of those exposed to the low BPA dose, all of the EB- and PPT-treated females, and none of the control animals. Ovaries from the EB-treated females were undersized and showed no evidence of folliculogenesis, whereas ovaries from the PPT-treated females were characterized by large antral-like follicles, which did not appear to support ovulation. Severity of deficits within the BPA-treated groups increased with dose and included large antral-like follicles and lower numbers of corpora lutea. Sexual receptivity, examined after ovariectomy and hormone replacement, was normal in all groups except those neonatally exposed to EB. FOS induction in hypothalamic gonadotropic (GnRH) neurons after hormone priming was impaired in the EB- and PPT-treated groups but neither of the BPA-treated groups. Our data suggest that BPA disrupts ovarian development but not the ability of GnRH neurons to respond to steroid-positive feedback.


Asunto(s)
Estrógenos no Esteroides/toxicidad , Hipotálamo/efectos de los fármacos , Ovario/efectos de los fármacos , Fenoles/toxicidad , Conducta Sexual Animal/efectos de los fármacos , Animales , Animales Recién Nacidos , Compuestos de Bencidrilo , Relación Dosis-Respuesta a Droga , Estrógenos no Esteroides/administración & dosificación , Ciclo Estral/efectos de los fármacos , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Masculino , Células Neuroendocrinas/efectos de los fármacos , Ovario/patología , Fenoles/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Long-Evans , Maduración Sexual/efectos de los fármacos
9.
Reprod Toxicol ; 27(2): 117-32, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19159674

RESUMEN

Genistein and ethinyl estradiol (EE(2)) were examined in multigenerational reproductive and chronic toxicity studies that had different treatment intervals among generations. Sprague-Dawley rats received genistein (0, 5, 100, or 500 ppm) or EE(2) (0, 2, 10, or 50 ppb) in a low phytoestrogen diet. Nonneoplastic effects in females are summarized here. Genistein at 500 ppm and EE(2) at 50 ppb produced similar effects in continuously exposed rats, including decreased body weights, accelerated vaginal opening, and altered estrous cycles in young animals. At the high dose, anogenital distance was subtly affected by both compounds, and a reduction in litter size was evident in genistein-treated animals. Genistein at 500 ppm induced an early onset of aberrant cycles relative to controls in the chronic studies. EE(2) significantly increased the incidence of uterine lesions (atypical focal hyperplasia and squamous metaplasia). These compound-specific effects appeared to be enhanced in the offspring of prior exposed generations.


Asunto(s)
Disruptores Endocrinos/toxicidad , Etinilestradiol/toxicidad , Genisteína/toxicidad , Reproducción/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Hiperplasia Endometrial/inducido químicamente , Hiperplasia Endometrial/patología , Estro/efectos de los fármacos , Femenino , Tamaño de la Camada/efectos de los fármacos , Metaplasia , Embarazo , Ratas , Ratas Sprague-Dawley , Conducta Sexual Animal/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Factores de Tiempo , Útero/efectos de los fármacos , Útero/patología , Vagina/efectos de los fármacos , Vagina/crecimiento & desarrollo
10.
Environ Health Perspect ; 116(3): 322-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18335098

RESUMEN

BACKGROUND: Although estrogenic chemicals can disrupt development of the reproductive system, there is debate about whether phytoestrogens in soy are beneficial, benign, or harmful. OBJECTIVES: We compared reproductive and metabolic characteristics in male and female mice reared and maintained on non-soy low-phytoestrogen feed or soy-based high-phytoestrogen feed. METHODS: The low-phytoestrogen diet was non-soy PMI 5K96 (verified casein diet), and the high-phytoestrogen diet consisted of soy-based PMI 5008 during pregnancy and lactation and soy-based PMI 5001 maintenance feed after weaning. RESULTS: In fetuses whose mothers consumed the low-phytoestrogen PMI 5K96 feed, we found a paradoxical significant elevation in endogenous serum estradiol, which was associated postnatally with adverse reproductive outcomes referred to as the "fetal estrogenization syndrome (FES)". In females, this syndrome included early puberty and increased uterine responsiveness to estrogen, and in males, it included reduced testis, epididymis, and seminal vesicle size, but an enlarged prostate. The low-phytoestrogen-fed males and females were lighter at birth, but, between weaning and adulthood, they became obese and developed abnormally high serum leptin levels; these males, but not females, showed impaired glucose regulation. CONCLUSIONS: Removing phytoestrogens from mouse feed produces an obese phenotype consistent with metabolic syndrome, and the associated reproductive system abnormalities are consistent with FES due to elevated endogenous fetal estradiol. Laboratory rodents may have become adapted to high-phytoestrogen intake over many generations of being fed soy-based commercial feed; removing all phytoestrogens from feed leads to alterations that could disrupt many types of biomedical research.


Asunto(s)
Estradiol/sangre , Genitales Femeninos/efectos de los fármacos , Genitales Masculinos/efectos de los fármacos , Intercambio Materno-Fetal , Obesidad/etiología , Fitoestrógenos/farmacología , Alimentación Animal , Animales , Animales Recién Nacidos , Peso al Nacer/efectos de los fármacos , Línea Celular Tumoral , Femenino , Genitales Femeninos/anatomía & histología , Genitales Masculinos/anatomía & histología , Prueba de Tolerancia a la Glucosa , Humanos , Leptina/sangre , Masculino , Exposición Materna , Ratones , Fitoestrógenos/administración & dosificación , Embarazo
11.
Int J Androl ; 31(2): 201-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18315718

RESUMEN

Environmental chemicals with hormone-like activity can disrupt the programming of endocrine signalling pathways that are established during perinatal life and result in adverse consequences that may not be apparent until much later in life. Increasing evidence implicates developmental exposure to environmental hormone mimics with a growing list of adverse health consequences in both males and females. Most recently, obesity has been proposed to be yet another adverse health effect of exposure to endocrine disrupting chemicals (EDCs) during critical stages of development. Obesity is quickly becoming a significant human health crisis because it is reaching epidemic proportions worldwide, and is associated with chronic illnesses such as diabetes and cardiovascular disease. In this review, we summarize the literature reporting an association of EDCs and the development of obesity, and further describe an animal model of exposure to diethylstilbestrol that has proven useful in studying mechanisms involved in abnormal programming of various oestrogen target tissues during differentiation. Together, these data suggest new targets (i.e. adipocyte differentiation and mechanisms involved in weight homeostasis) of abnormal programming by EDCs, and provide evidence that support the scientific term 'the developmental origins of adult disease'. The emerging idea of an association of EDCs and obesity expands the focus on obesity from intervention and treatment to include prevention and avoidance of these chemical modifiers.


Asunto(s)
Disruptores Endocrinos/toxicidad , Obesidad/inducido químicamente , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Feto/fisiopatología , Humanos , Masculino , Ratones
12.
Environ Int ; 115: 48-69, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29549716

RESUMEN

BACKGROUND: An increasing number of reports suggest early life exposures result in adverse effects in offspring who were never directly exposed; this phenomenon is termed "transgenerational inheritance." Given concern for public health implications for potential effects of exposures transmitted to subsequent generations, it is critical to determine how widespread and robust this phenomenon is and to identify the range of exposures and possible outcomes. OBJECTIVES: This scoping report examines the evidence for transgenerational inheritance associated with exposure to a wide range of stressors in humans and animals to identify areas of consistency, uncertainty, data gaps, and to evaluate general risk of bias issues for the transgenerational study design. METHODS: A protocol was developed to collect and categorize the literature into a systematic evidence map for transgenerational inheritance by health effects, exposures, and evidence streams following the Office of Health Assessment and Translation (OHAT) approach for conducting literature-based health assessments. RESULTS: A PubMed search yielded 63,758 unique records from which 257 relevant studies were identified and categorized into a systematic evidence map by evidence streams (46 human and 211 animal), broad health effect categories, and exposures. Data extracted from the individual studies are available in the Health Assessment Workspace Collaborative (HAWC) program. There are relatively few bodies of evidence where multiple studies evaluated the same exposure and the same or similar outcomes. Studies evaluated for risk of bias generally had multiple issues in design or conduct. CONCLUSIONS: The evidence mapping illustrated that risk of bias, few studies, and heterogeneity in exposures and endpoints examined present serious limitations to available bodies of evidence for assessing transgenerational effects. Targeted research is suggested to addressed inconsistencies and risk of bias issues identified, and thereby establish more robust bodies of evidence to critically assess transgenerational effects - particularly by adding data on exposure-outcome pairs where there is some evidence (i.e., reproductive, metabolic, and neurological effects).


Asunto(s)
Investigación Biomédica , Bases de Datos Factuales , Exposición a Riesgos Ambientales/análisis , Animales , Investigación Biomédica/métodos , Investigación Biomédica/normas , Femenino , Humanos , Masculino , Exposición Materna , Exposición Paterna , Embarazo , Efectos Tardíos de la Exposición Prenatal
13.
Endocrinology ; 148(8): 3580-90, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17446182

RESUMEN

In developing mouse ovaries, oocytes develop as clusters of cells called nests or germ cell cysts. Shortly after birth, oocyte nests dissociate and granulosa cells surround individual oocytes forming primordial follicles. At the same time, two thirds of the oocytes die by apoptosis, but the link between oocyte nest breakdown and oocyte death is unclear. Although mechanisms controlling breakdown of nests into individual oocytes and selection of oocytes for survival are currently unknown, steroid hormones may play a role. Treatment of neonatal mice with natural or synthetic estrogens results in abnormal multiple oocyte follicles in adult ovaries. Neonatal genistein treatment inhibits nest breakdown suggesting multiple oocyte follicles are nests that did not break down. Here we investigated the role of estrogen signaling in nest breakdown and oocyte survival. We characterized an ovary organ culture system that recapitulates nest breakdown, reduction in oocyte number, primordial follicle assembly, and follicle growth in vitro. We found that estradiol, progesterone, and genistein inhibit nest breakdown and primordial follicle assembly but have no effect on oocyte number both in organ culture and in vivo. Fetal ovaries, removed from their normal environment of high levels of pregnancy hormones, underwent premature nest breakdown and oocyte loss that was rescued by addition of estradiol or progesterone. Our results implicate hormone signaling in ovarian differentiation with decreased estrogen and progesterone at birth as the primary signal to initiate oocyte nest breakdown and follicle assembly. These findings also provide insight into the mechanism of multiple oocyte follicle formation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Estradiol/farmacología , Genisteína/farmacología , Oocitos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Progesterona/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Ratones , Ratones Endogámicos , Oocitos/fisiología , Técnicas de Cultivo de Órganos , Folículo Ovárico/citología , Folículo Ovárico/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
Mol Nutr Food Res ; 51(7): 832-44, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17604387

RESUMEN

Studies in our laboratory have shown that exposure to genistein causes deleterious effects on the developing female reproductive system. Mice treated neonatally on days 1-5 by subcutaneous injection of genistein (0.5-50 mg/kg) exhibited altered ovarian differentiation leading to multioocyte follicles (MOFs) at 2 months of age. Ovarian function and estrous cyclicity were also disrupted by neonatal exposure to genistein with increasing severity observed over time. Reduced fertility was observed in mice treated with genistein (0.5, 5, or 25 mg/kg) and infertility was observed at 50 mg/kg. Mammary gland and behavioral endpoints were also affected by neonatal genistein treatment. Further, transgenerational effects were observed; female offspring obtained from breeding genistein treated females (25 mg/kg) to control males had increased MOFs. Thus, neonatal treatment with genistein at environmentally relevant doses caused adverse consequences on female development which is manifested in adulthood. Whether adverse effects occur in human infants exposed to soy-based products such as soy infant formulas is unknown but the neonatal murine model may help address some of the current uncertainties since we have shown that many effects obtained from feeding genistin, the glycosolated form of genistein found in soy formula, are similar to those obtained from injecting genistein.


Asunto(s)
Genisteína/farmacología , Fitoestrógenos/farmacología , Reproducción/efectos de los fármacos , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Dieta , Ciclo Estral/efectos de los fármacos , Femenino , Genisteína/administración & dosificación , Genisteína/efectos adversos , Humanos , Recién Nacido , Masculino , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/crecimiento & desarrollo , Conducta Materna/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/crecimiento & desarrollo , Ovario/fisiología , Fitoestrógenos/administración & dosificación , Fitoestrógenos/efectos adversos , Reproducción/fisiología , Neoplasias Uterinas/inducido químicamente
15.
Mol Nutr Food Res ; 51(7): 912-7, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17604389

RESUMEN

Dietary substances and xenobiotic compounds with hormone-like activity can disrupt the programming of endocrine signaling pathways that are established during perinatal differentiation. The consequences of this disruption may not be apparent until later in life but increasing evidence implicates developmental exposure to environmental hormone-mimics with a growing list of adverse health effects including reproductive problems and increased cancer risks. Obesity has recently been proposed to be yet another adverse health consequence of exposure to endocrine disrupting substances during development. There is a renewed focus on identifying contributions of environmental factors to the development of obesity since it is reaching worldwide epidemic proportions, and this disease has the potential to overwhelm healthcare systems with associated illnesses such as diabetes and cardiovascular disease. Here, we review the literature that proposes an association of perinatal exposure to endocrine disrupting chemicals, in particular those with estrogenic activity, with the development of obesity later in life. We further describe an animal model of developmental exposure to diethylstilbestrol (DES) to study mechanisms involved in programming for obesity. Our experimental data support the idea that adipocytes and the mechanisms involved in weight homeostasis are novel targets of abnormal programming of environmental estrogens, some of which are found in our foods as naturally occurring substances or inadvertently as contaminants.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Estrógenos/toxicidad , Obesidad/etiología , Adipocitos , Envejecimiento , Animales , Dietilestilbestrol/toxicidad , Disruptores Endocrinos/toxicidad , Femenino , Edad Gestacional , Humanos , Masculino , Obesidad/epidemiología , Embarazo , Efectos Tardíos de la Exposición Prenatal
16.
Reprod Toxicol ; 23(3): 308-16, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17250991

RESUMEN

Studies in our laboratory have shown that developmental exposure to genistein causes deleterious effects on the reproductive system. Oral exposure to genistin (25mg/kg) increases uterine weight at 5 days of age similar to subcutaneous injection of genistein (20mg/kg) suggesting that subcutaneous injection of genistein is a suitable model for oral exposure to genistin. Mice treated neonatally by subcutaneous injection of genistein (0.5-50mg/kg) exhibit altered ovarian differentiation leading to multi-oocyte follicles (MOFs). Ovarian function and estrous cyclicity were disrupted in genistein treated mice with increasing severity over time. Reduced fertility was observed in mice treated with genistein (0.5, 5, or 25mg/kg) and infertility was observed at 50mg/kg. Females generated from genistein 25mg/kg females bred to control males have increased MOFs suggesting these effects can be transmitted to subsequent generations. Thus, neonatal treatment with genistein at environmentally relevant doses caused adverse consequences on reproduction in adulthood.


Asunto(s)
Genisteína/administración & dosificación , Genitales Femeninos/efectos de los fármacos , Fitoestrógenos/administración & dosificación , Reproducción/efectos de los fármacos , Animales , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Femenino , Genisteína/química , Genitales Femeninos/fisiopatología , Humanos , Estructura Molecular , Fitoestrógenos/química , Estilbenos/administración & dosificación
17.
Reprod Toxicol ; 24(2): 253-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17804194

RESUMEN

The developing fetus is uniquely sensitive to perturbation by chemicals with hormone-like activity. The adverse effects of prenatal diethylstilbestrol (DES) exposure are a classic example. Since concern has been mounting regarding the human health and environmental effects of bisphenol A (BPA), a high-production-volume chemical with estrogenic activity used in the synthesis of plastics, we investigated its long-term effects in an experimental animal model that was previously shown useful in studying the adverse effects of developmental exposure to DES. Outbred female CD-1 mice were treated on days 1-5 with subcutaneous injections of BPA (10, 100 or 1000 microg/kg/day) dissolved in corn oil or corn oil alone (Control). At 18 months, ovaries and reproductive tract tissues were examined. There was a statistically significant increase in cystic ovaries and cystic endometrial hyperplasia (CEH) in the BPA-100 group as compared to Controls. Progressive proliferative lesion (PPL) of the oviduct and cystic mesonephric (Wolffian) duct remnants were also seen in all of the BPA groups. More severe pathologies of the uterus following neonatal BPA treatment included adenomyosis, leiomyomas, atypical hyperplasia, and stromal polyps. These data suggest that BPA causes long-term adverse effects if exposure occurs during critical periods of differentiation.


Asunto(s)
Ovario/efectos de los fármacos , Fenoles/toxicidad , Adenocarcinoma/inducido químicamente , Adenocarcinoma/patología , Animales , Animales Recién Nacidos , Animales no Consanguíneos , Compuestos de Bencidrilo , Peso Corporal/efectos de los fármacos , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/patología , Relación Dosis-Respuesta a Droga , Hiperplasia Endometrial/inducido químicamente , Hiperplasia Endometrial/patología , Endometriosis/inducido químicamente , Endometriosis/patología , Estrógenos no Esteroides/administración & dosificación , Estrógenos no Esteroides/toxicidad , Femenino , Inyecciones Subcutáneas , Leiomioma/inducido químicamente , Leiomioma/patología , Masculino , Ratones , Quistes Ováricos/inducido químicamente , Quistes Ováricos/patología , Ovario/patología , Oviductos/efectos de los fármacos , Oviductos/patología , Fenoles/administración & dosificación , Embarazo , Factores de Tiempo , Neoplasias Uterinas/inducido químicamente , Neoplasias Uterinas/patología , Útero/efectos de los fármacos , Útero/patología
18.
Reprod Toxicol ; 23(3): 290-6, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17321108

RESUMEN

Xenobiotic and dietary compounds with hormone-like activity can disrupt endocrine signaling pathways that play important roles during perinatal differentiation and result in alterations that are not apparent until later in life. Evidence implicates developmental exposure to environmental hormone-mimics with a growing list of health problems. Obesity is currently receiving needed attention since it has potential to overwhelm health systems worldwide with associated illnesses such as diabetes and cardiovascular disease. Here, we review the literature that proposes an association of exposure to environmental endocrine disrupting chemicals with the development of obesity. We describe an animal model of developmental exposure to diethylstilbestrol (DES), a potent perinatal endocrine disruptor with estrogenic activity, to study mechanisms involved in programming an organism for obesity. This experimental animal model provides an example of the growing scientific field termed "the developmental origins of adult disease" and suggests new targets of abnormal programming by endocrine disrupting chemicals.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Disruptores Endocrinos/envenenamiento , Contaminantes Ambientales/envenenamiento , Obesidad/etiología , Animales , Dietilestilbestrol/envenenamiento , Disruptores Endocrinos/química , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/química , Femenino , Humanos , Obesidad/embriología , Obesidad/epidemiología , Embarazo
19.
Reprod Toxicol ; 24(2): 199-224, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17683900

RESUMEN

Concern is mounting regarding the human health and environmental effects of bisphenol A (BPA), a high-production-volume chemical used in synthesis of plastics. We have reviewed the growing literature on effects of low doses of BPA, below 50 mg/(kg day), in laboratory exposures with mammalian model organisms. Many, but not all, effects of BPA are similar to effects seen in response to the model estrogens diethylstilbestrol and ethinylestradiol. For most effects, the potency of BPA is approximately 10-1000-fold less than that of diethylstilbestrol or ethinylestradiol. Based on our review of the literature, a consensus was reached regarding our level of confidence that particular outcomes occur in response to low dose BPA exposure. We are confident that adult exposure to BPA affects the male reproductive tract, and that long lasting, organizational effects in response to developmental exposure to BPA occur in the brain, the male reproductive system, and metabolic processes. We consider it likely, but requiring further confirmation, that adult exposure to BPA affects the brain, the female reproductive system, and the immune system, and that developmental effects occur in the female reproductive system.


Asunto(s)
Fenoles/toxicidad , Conducta Sexual Animal/efectos de los fármacos , Animales , Compuestos de Bencidrilo , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/administración & dosificación , Disruptores Endocrinos/química , Disruptores Endocrinos/toxicidad , Femenino , Masculino , Ratones , Estructura Molecular , Fenoles/administración & dosificación , Fenoles/química , Ratas
20.
Endocrinology ; 147(10): 4871-82, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16857750

RESUMEN

Developmental effects of genistein (Gen) on the mammary gland were investigated using outbred female CD-1 mice treated neonatally on d 1-5 by sc injections at doses of 0.5, 5, or 50 mg/kg.d. Examination of mammary gland whole mounts (no. 4) before puberty (4 wk) revealed no morphological differences in development after Gen treatment. However, mice treated with Gen-50 had stunted development characterized by less branching at 5 wk and decreased numbers of terminal end buds at 5 and 6 wk. Conversely, at 6 wk, Gen-0.5-treated mice exhibited advanced development with increased ductal elongation compared with controls. Measurements of hormone receptor levels showed increased levels of progesterone receptor protein and estrogen receptor-beta mRNA in Gen-0.5-treated mice compared with controls; ERalpha expression was decreased after all doses of Gen treatment. Lactation ability, measured by pup weight gain and survival, was not affected after neonatal Gen-0.5 and Gen-5. Mice treated with Gen-50 did not deliver live pups; therefore, lactation ability could not be determined. Evaluation of mammary glands in aged mice (9 months) showed no differences between Gen-0.5-treated mice and controls but mice treated with Gen-5 and Gen-50 exhibited altered morphology including reduced lobular alveolar development, dilated ducts, and focal areas of "beaded" ducts lined with hyperplastic ductal epithelium. In summary, neonatal Gen exposure altered mammary gland growth and development as well as hormone receptor levels at all doses examined; higher doses of Gen led to permanent long-lasting morphological changes.


Asunto(s)
Animales Recién Nacidos/fisiología , Genisteína/farmacología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Fitoestrógenos/farmacología , Animales , Peso Corporal/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Hormonas/sangre , Inmunohistoquímica , Lactancia/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Ratones , Embarazo , Receptores de Prolactina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sobrevida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA