Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Total Environ ; 805: 150371, 2022 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-34818814

RESUMEN

Progestins and estrogens are widespread in various aquatic environments and their potential endocrine disruption effects to aquatic organisms have drawn growing concern. However, their combined effects in aquatic organisms remain elusive. The aim of the present study was to assess the effects of the binary mixtures of gestodene (GES) and 17α-ethinylestradiol (EE2) on the hypothalamic-pituitary-thyroid (HPT) axis of zebrafish (Danio rerio) using the eleuthero-embryos. Embryos were exposed to GES and EE2 alone or in combination at concentrations ranging from 41 to 5329 ng L-1 (nominal ones from 50 to 5000 ng L-1) for 48 h, 96 h and 144 h post fertilization (hpf). The results showed that the transcripts of the genes along the HPT axis displayed pronounced alterations. There was no clear pattern in the change of the transcripts of these genes over time and with concentrations. However, in general, the transcripts of the genes were inversely affected by EE2 (increase 0.5 to 4.2-folds) and GES (inhibition 0.4 to 4.9-folds), and their mixtures showed interactive effects in embryonic zebrafish. In addition, physiological data (mortality, malformation, body length and heart rate etc.) denoted higher toxicity of the two chemicals in combination than alone based on the developmental toxicity and neurotoxicity (locomotor behavior). These results indicated that the interactive effects of these two chemicals might be different between at the transcriptional level and at the whole organismal level. In summary, GES and EE2 affect the HPT axis (related genes expression and thyroid hormones (THs) levels) and exhibit developmental toxicity and neurotoxicity.


Asunto(s)
Contaminantes Químicos del Agua , Pez Cebra , Animales , Etinilestradiol/toxicidad , Hipotálamo , Progesterona , Glándula Tiroides , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/genética
2.
Neurotoxicology ; 74: 139-148, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31251963

RESUMEN

Bisphenol A (BPA) is a well-characterized endocrine disrupting chemical (EDC) used in plastics, epoxy resins and other products. Neurodevelopmental effects of BPA exposure are a major concern with multiple rodent and human studies showing that early life BPA exposure may impact the developing brain and sexually dimorphic behaviors. The CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity) program was established to assess multiple endpoints, including neural, across a wide dose range. Studies from our lab as part of (and prior to) CLARITY-BPA have shown that BPA disrupts estrogen receptor expression in the developing brain, and some evidence of oxytocin (OT) and oxytocin receptor (OTR) disruption in the hypothalamus and amygdala. While BPA disruption of steroid hormone function is well documented, less is known about its capacity to alter nonapeptide signals. In this CLARITY-BPA follow up study, we used remaining juvenile rat tissues to test the hypothesis that developmental BPA exposure affects OTR expression across the brain. Perinatal BPA exposure (2.5, 25, or 2500 µg/kg body weight (bw)/day) spanned gestation and lactation with dams gavaged from gestational day 6 until birth and then the offspring gavaged directly through weaning. Ethinyl estradiol (0.5 µg/kg bw/day) was used as a reference estrogen. Animals of both sexes were sacrificed as juveniles and OTR expression assessed by receptor binding. Our results demonstrate prenatal exposure to BPA can eliminate sex differences in OTR expression in three hypothalamic regions, and that male OTR expression may be more susceptible. Our data also identify a sub-region of the BNST with sexually dimorphic OTR expression not previously reported in juvenile rats that is also susceptible to BPA.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Química Encefálica/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Receptores de Oxitocina/biosíntesis , Animales , Animales Recién Nacidos , Etinilestradiol/toxicidad , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Oxitocina/biosíntesis , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Caracteres Sexuales
3.
Environ Toxicol Chem ; 38(3): 533-547, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30569562

RESUMEN

Fish (embryo) toxicity test guidelines are mostly based on aquatic exposures. However, in some cases, other exposure routes can be more practical and relevant. Micro-injection into the yolk of fish embryos could offer a particular advantage for administering hydrophobic compounds, such as many endocrine disruptors. Single-dose micro-injection was compared with continuous aquatic exposure in terms of compound accumulation and biological responses. 17α-Ethinyl estradiol (EE2) was used as a model compound. First, the optimal solvent and droplet size were optimized, and needle variation was assessed. Next, biological endpoints were evaluated. The accumulated internal dose of EE2 decreased over time in both exposure scenarios. Estrogen receptor activation was concentration/injected dose dependent, increased daily, and was related to esr2b transcription. Transcription of vitellogenin 1 (vtg1) and brain aromatase (cyp19a1b) was induced in both scenarios, but the cyp19a1b transcription pattern differed between routes. Injection caused an increase in cyp19a1b transcripts from 48 hours post fertilization (hpf) onward, whereas after aquatic exposure the main increase occurred between 96 and 120 hpf. Some malformations only occurred after injection, whereas others were present for both scenarios. We conclude that responses can differ between exposure routes and therefore micro-injection is not a direct substitute for, but can be complementary to aquatic exposure. Nevertheless, vtg1and cyp19a1b transcription and estrogen receptor activation are suitable biomarkers for endocrine disruptor screening in both scenarios. Environ Toxicol Chem 2019;38:533-547. © 2018 SETAC.


Asunto(s)
Disruptores Endocrinos/administración & dosificación , Pruebas de Toxicidad/métodos , Contaminantes Químicos del Agua/administración & dosificación , Animales , Aromatasa/genética , Aromatasa/metabolismo , Embrión no Mamífero/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Etinilestradiol/administración & dosificación , Etinilestradiol/toxicidad , Masculino , Microinyecciones/métodos , Receptores de Estrógenos/metabolismo , Vitelogeninas/genética , Vitelogeninas/metabolismo , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
J Tradit Chin Med ; 39(3): 402-409, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-32186012

RESUMEN

OBJECTIVE: To investigate the pathway through which Calculus Bovis Sativus (CBS) up-regulates hepatic multidrug resistance-associated protein 2 (Mrp2) and Mrp4 in 17α-ethynylestradiol (EE)-induced cholestasis. METHODS: Five groups of rats were designed: control group, EE+ICI182780 group, EE group, EE+CBS 50 mg/kg group and EE + CBS 150 mg/kg group. CBS (50 and 150 mg·kg-1·d-1 ) was orally given to rats by gavage for five consecutive days in coadministration with EE. The levels of cholestasis biomarkers, alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) and total bilirubin (TBIL) were determined by biochemical methods. The bile flow was measured. The histopathology of the liver tissue was evaluated. The expression of Mrp2, Mrp3, Mrp4, estrogen receptor α (ERα) and ERß was determined by Western blotting. RESULTS: CBS markedly improved EE-induced cholestasis. EE exposure significantly reduced hepatic Mrp2 and Mrp4 expression compared with the control group. EE also dramatically up-regulated the expression of Mrp3. Compared to the EE group, CBS notably up-regulated hepatic Mrp2 and Mrp4 but failed to influence the Mrp3 level significantly. ICI182780, an ER antagonist, showed similar beneficial effects as CBS. Decreased expression of Mrp2 and Mrp4 caused by EE was also restored by ICI182780. Additionally, EE significantly induced he- patic ERα expression, which was reversed by ICI182780 or CBS (150 mg/kg) treatment, suggesting that CBS exerted a moderate regulatory effect on ER signaling. CONCLUSION: CBS up-regulated hepatic Mrp2 and Mrp4 expression in EE-induced cholestasis, which might be associated with its regulation of ER signaling.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Colestasis/inducido químicamente , Colestasis/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Etinilestradiol/toxicidad , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Animales , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
5.
Curr Med Sci ; 38(1): 167-173, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30074167

RESUMEN

Baicalin is one of the main active ingredients of choleretic traditional Chinese medicine drug Radix Scutellariae. The aim of this study was to explore the pharmacokinetic characteristics of baicalin in rats with 17α-ethynylestradiol (EE)-induced intrahepatic cholestasis (IC) based on its choleretic effects. Firstly, rats were subcutaneously injected with EE solution (5 mg/kg, 0.25 mL/100 g) for 5 consecutive days to construct an IC model. Then the bile excretion rate, serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) and total bile acid (TBA) and pathological changes of the liver were detected. Secondly, after successfully modeling, the rats were intragastrically given baicalin solution (200 mg/kg) (n=6). Blood samples were collected from the tail vein at different time points after intragastric administration. The protective effects of low- (50 mg/kg), medium- (100 mg/kg) and high-dose (200 mg/kg) baicalin on the liver in IC rats were evaluated. The content of baicalin in plasma was detected by liquid chromatography-mass spectrometry/mass spectrometry and pharmacokinetics parameters were calculated. Pharmacodynamic results showed that low-, medium- and high-dose baicalin all significantly increased the average excretion rate of bile (P<0.05), and significantly decreased serum levels of ALT, AST and ALP and TBA (P<0.05). Meanwhile, HE staining showed that baicalin significantly relieved EE-induced hepatocyte edema and necrosis. Pharmacokinetic results exhibited that the absorption of baicalin in both IC and normal control rats showed bimodal phenomenon. Cmax, AU(0-t) and AUC(0-∞) of baicalin in IC rats were significantly higher than those of the normal control group (P<0.01). T1/2 of plasma baicalin in the model group was significantly extended to (11.09±1.84) h, with clearance dropping to 61.78% of that of the normal control group (P<0.01). The above results suggested that baicalin had protective effects on the liver of IC rats, accompanied by significantly increased in vivo exposure, delayed in vivo clearance and markedly alterative pharmacokinetic characteristics. This study provides a theoretical basis for further development of baicalin as a feasible drug for treating IC.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Colagogos y Coleréticos/farmacocinética , Colestasis Intrahepática/tratamiento farmacológico , Flavonoides/farmacocinética , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Colagogos y Coleréticos/administración & dosificación , Colagogos y Coleréticos/uso terapéutico , Colestasis Intrahepática/etiología , Etinilestradiol/toxicidad , Flavonoides/administración & dosificación , Flavonoides/uso terapéutico , Masculino , Ratas , Ratas Wistar
6.
Toxicol Sci ; 160(1): 15-29, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973534

RESUMEN

Current strategies for predicting carcinogenic mode of action for nongenotoxic chemicals are based on identification of early key events in toxicity pathways. The goal of this study was to evaluate short-term key event indicators resulting from exposure to androstenedione (A4), an androgen receptor agonist and known liver carcinogen in mice. Liver cancer is more prevalent in men compared with women, but androgen-related pathways underlying this sex difference have not been clearly identified. Short-term hepatic effects of A4 were compared with reference agonists of the estrogen receptor (ethinyl estradiol, EE) and glucocorticoid receptor (prednisone, PRED). Male B6C3F1 mice were exposed for 7 or 28 days to A4, EE, or PRED. EE increased and PRED suppressed hepatocyte proliferation, while A4 had no detectable effects. In a microarray analysis, EE and PRED altered >3000 and >670 genes, respectively, in a dose-dependent manner, whereas A4 did not significantly alter any genes. Gene expression was subsequently examined in archival liver samples from male and female B6C3F1 mice exposed to A4 for 90 days. A4 altered more genes in females than males and did not alter expression of genes linked to activation of the mitogenic xenobiotic receptors AhR, CAR, and PPARα in either sex. A gene expression biomarker was used to show that in female mice, the high dose of A4 activated the growth hormone-regulated transcription factor STAT5b, which controls sexually dimorphic gene expression in the liver. These findings suggest that A4 induces subtle age-related effects on STAT5b signaling that may contribute to the higher risk of liver cancer in males compared with females.


Asunto(s)
Androstenodiona/toxicidad , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/química , Transformación Celular Neoplásica/genética , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/genética , Hígado/efectos de los fármacos , Animales , Biomarcadores de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Etinilestradiol/toxicidad , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Fenotipo , Prednisona/toxicidad , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Factores Sexuales , Factores de Tiempo , Transcriptoma
7.
Physiol Rep ; 5(3)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28196854

RESUMEN

Bisphenol A (BPA) is an endocrine-disrupting chemical (EDC) prevalent in many household items. Rodent models and human epidemiological studies have linked this chemical to neurobehavior impairments. In California mice, developmental exposure to BPA results in sociosexual disorders at adulthood, including communication and biparental care deficits, behaviors that are primarily regulated by the hypothalamus. Thus, we sought to examine the transcriptomic profile in this brain region of juvenile male and female California mice offspring exposed from periconception through lactation to BPA or ethinyl estradiol (EE, estrogen present in birth control pills and considered a positive estrogen control for BPA studies). Two weeks prior to breeding, P0 females were fed a control diet, or this diet supplemented with 50 mg BPA/kg feed weight or 0.1 ppb EE, and continued on the diets through lactation. At weaning, brains from male and female offspring were collected, hypothalamic RNA isolated, and RNA-seq analysis performed. Results indicate that BPA and EE groups clustered separately from controls with BPA and EE exposure leading to unique set of signature gene profiles. Kcnd3 was downregulated in the hypothalamus of BPA- and EE-exposed females, whereas Tbl2, Topors, Kif3a, and Phactr2 were upregulated in these groups. Comparison of transcripts differentially expressed in BPA and EE groups revealed significant enrichment of gene ontology terms associated with microtubule-based processes. Current results show that perinatal exposure to BPA or EE can result in several transcriptomic alterations, including those associated with microtubule functions, in the hypothalamus of California mice. It remains to be determined whether these genes mediate BPA-induced behavioral disruptions.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Etinilestradiol/toxicidad , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Regulación hacia Abajo , Femenino , Masculino , Exposición Materna , Peromyscus , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Regulación hacia Arriba
8.
Biol Reprod ; 93(2): 32, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26134866

RESUMEN

Neonatal exposure to estrogens is known to cause delayed effects, a late-occurring adverse effect on adult female reproductive functions, such as early onset of age-matched abnormal estrous cycling. However, the critical period in which neonates are sensitive to delayed effects inducible by exogenous estrogen exposure has not been clearly identified. To clarify this window, we examined the intensity and timing of delayed effects using rats exposed to ethynylestradiol (EE) at various postnatal ages. After subcutaneous administration of a single dose of EE (20 µg/kg, which induces delayed effects) on Postnatal Day (PND) 0, 5, 10, or 14 in Wistar rats, hypothalamic and hormonal alterations in young adults and long-term estrous cycling status were investigated as indicators of delayed effects. In young adults, peak luteinizing hormone concentrations at the time of the luteinizing hormone surge showed a decreasing trend, and KiSS1 mRNA expression of the anterior hypothalamus and number of KiSS1-positive cells in the anteroventral periventricular nucleus were significantly decreased in the PND 0, 5, and 10 groups. The reduction in KiSS1 mRNA and KiSS1-postive cells was inversely correlated with age at time of exposure. These groups also exhibited early onset of abnormal estrous cycling, starting from 17 wk of age in the PND0 group and 19 wk of age in the PND5 and 10 groups. These indicators were not apparent in the PND14 group. Our results suggest that PND0-PND10 is the critical window of susceptibility for delayed effects, and PND14 is presumed to be the provisional endpoint of the window.


Asunto(s)
Etinilestradiol/toxicidad , Envejecimiento , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Ciclo Estral/efectos de los fármacos , Etinilestradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Hipotálamo/efectos de los fármacos , Hipotálamo/crecimiento & desarrollo , Hipotálamo Anterior/metabolismo , Kisspeptinas/biosíntesis , Kisspeptinas/genética , Hormona Luteinizante/sangre , Embarazo , Efectos Tardíos de la Exposición Prenatal , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Diferenciación Sexual/efectos de los fármacos , Vagina/efectos de los fármacos , Vagina/crecimiento & desarrollo , Enfermedades Vaginales/inducido químicamente , Enfermedades Vaginales/patología
9.
Psychoneuroendocrinology ; 54: 1-13, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25679306

RESUMEN

Ethinyl Estradiol (EE), a synthetic, orally bio-available estrogen, is the most commonly prescribed form of estrogen in oral contraceptives, and is found in at least 30 different contraceptive formulations currently prescribed to women as well as hormone therapies prescribed to menopausal women. Thus, EE is prescribed clinically to women at ages ranging from puberty to reproductive senescence. Here, in two separate studies, the cognitive effects of cyclic or tonic EE administration following ovariectomy (Ovx) were evaluated in young female rats. Study I assessed the cognitive effects of low and high doses of EE, delivered tonically via a subcutaneous osmotic pump. Study II evaluated the cognitive effects of low, medium, and high doses of EE administered via a daily subcutaneous injection, modeling the daily rise and fall of serum EE levels with oral regimens. Study II also investigated the impact of low, medium and high doses of EE on the basal forebrain cholinergic system. The low and medium doses utilized here correspond to the range of doses currently used in clinical formulations, and the high dose corresponds to doses prescribed to a generation of women between 1960 and 1970, when oral contraceptives first became available. We evaluate cognition using a battery of maze tasks tapping several domains of spatial learning and memory as well as basal forebrain cholinergic integrity using immunohistochemistry and unbiased stereology to estimate the number of choline acetyltransferase (ChAT)-producing cells in the medial septum and vertical/diagonal bands. At the highest dose, EE treatment impaired multiple domains of spatial memory relative to vehicle treatment, regardless of administration method. When given cyclically at the low and medium doses, EE did not impact working memory, but transiently impaired reference memory during the learning phase of testing. Of the doses and regimens tested here, only EE at the highest dose impaired several domains of memory; tonic delivery of low EE, a dose that corresponds to the most popular doses used in the clinic today, did not impact cognition on any measure. Both medium and high injection doses of EE reduced the number of ChAt-immunoreactive cells in the basal forebrain, and cell population estimates in the vertical/diagonal bands negatively correlated with working memory errors.


Asunto(s)
Prosencéfalo Basal/efectos de los fármacos , Neuronas Colinérgicas/efectos de los fármacos , Cognición/efectos de los fármacos , Etinilestradiol/toxicidad , Trastornos de la Memoria/inducido químicamente , Animales , Anticonceptivos Orales/administración & dosificación , Anticonceptivos Orales/toxicidad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Etinilestradiol/administración & dosificación , Femenino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Endogámicas F344
10.
Toxicol Sci ; 140(1): 190-203, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24752507

RESUMEN

Concerns have been raised regarding the long-term impacts of early life exposure to the ubiquitous environmental contaminant bisphenol A (BPA) on brain organization. Because BPA has been reported to affect estrogen signaling, and steroid hormones play a critical role in brain sexual differentiation, there is also concern that BPA exposure could alter neural sex differences. Here, we examine the impact of subchronic exposure from gestation to adulthood to oral doses of BPA below the current no-observed-adverse-effect level (NOAEL) of 5 mg/kg body weight (bw)/day on estrogen receptor (ESR) expression in sexually dimorphic brain regions of prepubertal and adult female rats. The dams were gavaged daily with vehicle (0.3% carboxymethylcellulose), 2.5, 25, 260, or 2700 µg BPA/kg bw/day, or 0.5 or 5.0 µg ethinyl estradiol (EE)/kg bw/day from gestational day 6 until labor began. Offspring were then gavaged directly from the day after birth until the day before scheduled sacrifice on postnatal days 21 or 90. Using in situ hybridization, one or more BPA doses produced significant decreases in Esr1 expression in the juvenile female rat anteroventral periventricular nucleus (AVPV) of the hypothalamus and significant decreases in Esr2 expression in the adult female rat AVPV and medial preoptic area (MPOA), relative to vehicle controls. BPA did not simply reproduce EE effects, indicating that BPA is not acting solely as an estrogen mimic. The possible consequences of long-term changes in hypothalamic ESR expression resulting from subchronic low dose BPA exposure on neuroendocrine effects are discussed and being addressed in ongoing, related work.


Asunto(s)
Envejecimiento , Compuestos de Bencidrilo/toxicidad , Etinilestradiol/toxicidad , Hipotálamo/efectos de los fármacos , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Receptores de Estrógenos/genética , Envejecimiento/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Expresión Génica/efectos de los fármacos , Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Sprague-Dawley , Caracteres Sexuales
11.
Neurotoxicology ; 37: 154-62, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23660487

RESUMEN

Early life exposure to endocrine disruptors is considered to disturb normal development of hormone sensitive parameters and contribute to advanced puberty and reduced fecundity in humans. Kisspeptin is a positive regulator of the hypothalamic-pituitary-gonadal axis, and plays a key role in the initiation of puberty. In the adult, Kiss1 gene expression occurs in two hypothalamic nuclei, namely the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC), which are differentially regulated by peripheral sex steroid hormones. In this study we determined the effects on puberty onset and Kiss1 mRNA levels in each of the two nuclei after long-term perinatal exposure of rats to ethinyl oestradiol (EE2) or to five different pesticides, individually and in a mixture. Rat dams were per orally administered with three doses of EE2 (5, 15 or 50 µg/kg/day) or with the pesticides epoxiconazole, mancozeb, prochloraz, tebuconazole, and procymidone, alone or in a mixture of the five pesticides at three different doses. Kiss1 mRNA expression was determined in the AVPV and in the ARC of the adult male and female pups in the EE2 experiment, and in the adult female pups in the pesticide experiment. We find that perinatal EE2 exposure did not affect Kiss1 mRNA expression in this study designed to model human exposure to estrogenic compounds, and we find only minor effects on puberty onset. Further, the Kiss1 system does not exhibit persistent changes and puberty onset is not affected after perinatal exposure to a pesticide mixture in this experimental setting. However, we find that the pesticide mancozeb tends to increase Kiss1 expression in the ARC, presumably through neurotoxic mechanisms rather than via classical endocrine disruption, calling for increased awareness that Kiss1 expression can be affected by environmental pollutants through multiple mechanisms.


Asunto(s)
Disruptores Endocrinos/toxicidad , Etinilestradiol/toxicidad , Hipotálamo/efectos de los fármacos , Kisspeptinas/metabolismo , Neuronas/efectos de los fármacos , Plaguicidas/toxicidad , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Femenino , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Kisspeptinas/genética , Masculino , Neuronas/metabolismo , Embarazo , ARN Mensajero/metabolismo , Ratas , Maduración Sexual
12.
PLoS One ; 8(1): e54655, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349947

RESUMEN

BACKGROUND: The Ayurvedic medicinal system claims Mucuna pruriens (MP) to possess pro-male fertility, aphrodisiac and adaptogenic properties. Some scientific evidence also supports its pro-male fertility properties; however, the mechanism of its action is not yet clear. The present study aimed at demonstrating spermatogenic restorative efficacy of MP and its major constituent L-DOPA (LD), and finding the possible mechanism of action thereof in a rat model. METHODOLOGY/FINDINGS: Ethinyl estradiol (EE) was administered at a rate of 3 mg/kg body weight (BW)/day for a period of 14 days to generate a rat model with compromised spermatogenesis. MP and LD were administered in two separate groups of these animals starting 15(th) day for a period of 56 days, and the results were compared with an auto-recovery (AR) group. Sperm count and motility, testis histo-architecture, level of reactive oxygen species (ROS), mitochondrial membrane potential (MMP), apoptosis, peripheral hormone levels and testicular germ cell populations were analysed, in all experimental groups. We observed efficient and quick recovery of spermatogenesis in MP and LD groups in comparison to the auto-recovery group. The treatment regulated ROS level, apoptosis, and mitochondrial membrane potential (MMP), recovered the hypothalamic-pituitary-gonadal axis and the number of testicular germ cells, ultimately leading to increased sperm count and motility. CONCLUSION/SIGNIFICANCE: M. pruriens efficiently recovers the spermatogenic loss induced due to EE administration. The recovery is mediated by reduction in ROS level, restoration of MMP, regulation of apoptosis and eventual increase in the number of germ cells and regulation of apoptosis. The present study simplified the complexity of mechanism involved and provided meaningful insights into MP/LD mediated correction of spermatogenic impairment caused by estrogens exposure. This is the first study demonstrating that L-DOPA largely accounts for pro-spermatogenic properties of M. pruriens. The manuscript bears CDRI communication number 8374.


Asunto(s)
Fármacos para la Fertilidad Masculina/administración & dosificación , Infertilidad Masculina/tratamiento farmacológico , Infertilidad Masculina/fisiopatología , Levodopa/administración & dosificación , Extractos Vegetales/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Etinilestradiol/toxicidad , Humanos , Infertilidad Masculina/inducido químicamente , Masculino , Medicina Ayurvédica , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mucuna/química , Extractos Vegetales/química , Ratas , Especies Reactivas de Oxígeno/metabolismo , Motilidad Espermática/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatozoides/citología , Espermatozoides/efectos de los fármacos , Espermatozoides/crecimiento & desarrollo
13.
J Toxicol Sci ; 37(4): 681-90, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22863849

RESUMEN

Delayed effects of exposure to small amounts of estrogenic compounds during the critical period of brain sexual differentiation were investigated by subcutaneous treatment of female Sprague-Dawley rats with 0 (vehicle control), 0.08, 0.4, or 2 µg/kg of 17α-ethynylestradiol (EE) on postnatal day (PND) 1. The treatment did not affect growth and development of the treated animals, and the timings of vaginal opening were similar between the EE-treated and control groups. The animals were periodically examined for the estrous cycle from postnatal week (PNW) 8-9 to PNW 32-33. Patterns of the estrous cycle were similar among the groups until PNW 17. None of the control animals showed persistent estrus until PNW 33. The animals treated with 0.4 µg/kg or more EE showed persistent estrus from PNW 20. The alteration was reflected in the number of days judged as proestrus or estrus, and was found to gradually increase in the EE-treated groups. At necropsy on PNW 32-33, ovulation was not confirmed in most EE-treated animals, even on the day of estrus. In addition, sporadic milk accumulations were observed in the mammary gland of the EE-treated animals. Histological evaluation revealed cystic follicle formation in the EE-treated ovaries and also revealed hyperplasia of mammary glands. Furthermore, ovaries from the animals showing persistent estrus lacked corpus luteum, indicating long-term anovulation. These results clearly show that single exposure to EE during the critical period of brain sexual differentiation can exert effects on reproductive functions at a later period in rats.


Asunto(s)
Etinilestradiol/toxicidad , Ovulación/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Estro/efectos de los fármacos , Femenino , Tamaño de los Órganos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Diferenciación Sexual/efectos de los fármacos , Vagina/efectos de los fármacos
14.
Environ Sci Technol ; 46(6): 3472-9, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22360147

RESUMEN

Environmental estrogens have been shown to affect aspects of fish behavior that could potentially impact on wild populations, but the physiological mechanisms underpinning these effects are unknown. Using small colonies of zebrafish (Danio rerio), we evaluated the impacts of estrogen exposure on the aggression of dominant males, the associated implications for their social status and reproductive success, and their signaling mechanisms. The aggression of dominant males exposed to 17α-ethinylestradiol (EE(2); 10 ng/L nominal) was reduced significantly, and half of these fish subsequently lost their dominance, behavioral changes that were reflected in their reproductive success. Plasma androgen and the expression of genes involved in sex steroid production/signaling (cyp19a1b, cyp17, hsd11b2, hsd17b3, ar) and aggression (avplrv1b, tph1b, htr1a, sst1, sstr1, th, slc6a3, ar) were higher in control dominant versus subordinate males, but suppressed by EE(2) exposure, such that the differences between the social ranks were not retained. The expression levels of avpl (brain), which promotes aggression and dominance, and ar and cyp17 (gonad) were elevated in nonexposed males paired with EE(2)-exposed males. Our findings illustrate that disruptions of behaviors affecting social hierarchy, and in turn breeding outcome, as a consequence of exposure to an environmental estrogen are signaled through complex interconnecting gonadal and neurological control mechanisms that generally conform with those established in mammalian models. The extensive molecular, genetic, physiological, and behavioral toolbox now available for the zebrafish makes this species an attractive model for integrated analyses of chemical effects spanning behavior to molecular effect mechanisms.


Asunto(s)
Estrógenos/toxicidad , Etinilestradiol/toxicidad , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/fisiología , Agresión/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Femenino , Proteínas de Peces/genética , Regulación de la Expresión Génica/efectos de los fármacos , Gónadas/efectos de los fármacos , Gónadas/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Reproducción/efectos de los fármacos , Predominio Social , Testosterona/análogos & derivados , Testosterona/sangre
15.
Fish Physiol Biochem ; 38(3): 653-64, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21850400

RESUMEN

Endocrine-disrupting chemicals (EDCs) can affect normal sexual differentiation in fish. Foxl2, one forkhead transcription factor, plays an important role in ovarian differentiation in the early development of the female gonad in mammals and fish. How EDCs affect Foxl2 expression is little known. In this study, we isolated a Foxl2 cDNA from the ovary of rare minnow Gobiocypris rarus and examined its expression during early development stages and in different adult tissues. Then, we analyzed Foxl2 expression in G. rarus juvenile following 3-day exposure to 17α- ethinylestradiol (EE2), 4-n-nonylphenol (NP), and bisphenol A (BPA). Alignment of known Foxl2 sequences among vertebrates showed high identity in forkhead domain and C-terminal region with other vertebrate proteins. Quantitative RT-PCR analysis showed that Foxl2 expression was linear decrease and cyp19a1a, the downstream target gene of Foxl2, had no correlation with Foxl2 from 18 to 50 days post fertilization (dpf). Among different adult tissues, Foxl2 is mainly expressed in ovary, brain, gill, eye, and male spleen. In the 3-day exposure, the juvenile fish to EDCs, 0.1 nM EE2, and 1 nM BPA significantly up-regulated the expression of Foxl2 gene, while NP had no effect on Foxl2 expression. Altogether, these results provide basic data for further study on how Foxl2 mediates EDCs impact on the sexual differentiation in G. rarus.


Asunto(s)
Cyprinidae/genética , Cyprinidae/metabolismo , Disruptores Endocrinos/toxicidad , Proteínas de Peces/genética , Factores de Transcripción Forkhead/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencia de Aminoácidos , Animales , Aromatasa/genética , Secuencia de Bases , Compuestos de Bencidrilo , Clonación Molecular , Cyprinidae/crecimiento & desarrollo , ADN Complementario/genética , Etinilestradiol/toxicidad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Datos de Secuencia Molecular , Ovario/efectos de los fármacos , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Fenoles/toxicidad , Filogenia , Reacción en Cadena en Tiempo Real de la Polimerasa , Homología de Secuencia de Aminoácido , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/genética , Distribución Tisular , Regulación hacia Arriba/efectos de los fármacos
16.
Reprod Toxicol ; 28(3): 342-53, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19383540

RESUMEN

Genistein and ethinyl estradiol (EE(2)) were examined in multigenerational reproductive and 2-yr chronic toxicity studies with different exposure durations across generations F(0) through F(4). Sprague-Dawley rats were exposed to genistein (0, 5, 100, or 500 ppm) or EE(2) (0, 2, 10, or 50 ppb). Effects in the male mammary gland are described here. In the multigeneration studies, mammary hyperplasia was induced by both compounds; the chronic studies had a lower incidence, without proportionate neoplasia. Sexual dimorphism (predominant tubuloalveolar growth in females and lobuloalveolar in males) was retained without feminization in high dose genistein or EE(2). In the continuously exposed generations, mammary hyperplasia was sustained but not amplified, appeared morphologically similar across all generations, and was not carried over into unexposed offspring of previously exposed generations. The hyperplasia in male rats was similar whether induced by genistein or EE(2). Results substantiate and extend previous reports that mammary gland hyperplasia in the male rat is one of the most sensitive markers of estrogenic endocrine disruption.


Asunto(s)
Estrógenos/toxicidad , Etinilestradiol/toxicidad , Genisteína/toxicidad , Glándulas Mamarias Animales/efectos de los fármacos , Fitoestrógenos/toxicidad , Reproducción/efectos de los fármacos , Administración Oral , Alimentación Animal , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Hiperplasia/inducido químicamente , Masculino , Glándulas Mamarias Animales/patología , Exposición Materna/efectos adversos , Embarazo , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad Crónica
17.
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
18.
Toxicol Sci ; 102(1): 42-60, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18084045

RESUMEN

Among proposed uses for microarrays in environmental toxiciology is the identification of key contributors to toxicity within a mixture. However, it remains uncertain whether the transcriptomic profiles resulting from exposure to a mixture have patterns of altered gene expression that contain identifiable contributions from each toxicant component. We exposed isogenic rainbow trout Onchorynchus mykiss, to sublethal levels of ethynylestradiol, 2,2,4,4-tetrabromodiphenyl ether, and chromium VI or to a mixture of all three toxicants Fluorescently labeled complementary DNA (cDNA) were generated and hybridized against a commercially available Salmonid array spotted with 16,000 cDNAs. Data were analyzed using analysis of variance (p<0.05) with a Benjamani-Hochberg multiple test correction (Genespring [Agilent] software package) to identify up and downregulated genes. Gene clustering patterns that can be used as "expression signatures" were determined using hierarchical cluster analysis. The gene ontology terms associated with significantly altered genes were also used to identify functional groups that were associated with toxicant exposure. Cross-ontological analytics approach was used to assign functional annotations to genes with "unknown" function. Our analysis indicates that transcriptomic profiles resulting from the mixture exposure resemble those of the individual contaminant exposures, but are not a simple additive list. However, patterns of altered genes representative of each component of the mixture are clearly discernible, and the functional classes of genes altered represent the individual components of the mixture. These findings indicate that the use of microarrays to identify transcriptomic profiles may aid in the identification of key stressors within a chemical mixture, ultimately improving environmental assessment.


Asunto(s)
Monitoreo del Ambiente/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncorhynchus mykiss/genética , Contaminantes Químicos del Agua/toxicidad , Animales , Carga Corporal (Radioterapia) , Cromo/toxicidad , Análisis por Conglomerados , Interacciones Farmacológicas , Etinilestradiol/toxicidad , Éteres Difenilos Halogenados , Hidrocarburos Bromados/toxicidad , Hígado/metabolismo , Éteres Fenílicos/toxicidad , Reacción en Cadena de la Polimerasa , Reproducibilidad de los Resultados , Factores de Tiempo , Contaminantes Químicos del Agua/farmacocinética
19.
Environ Pollut ; 142(3): 559-66, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16413089

RESUMEN

This study evaluated the effect of aquatic secondary nutrient supply levels (nitrogen and phosphorus) on the subcellular response of adult male fathead minnows (Pimephales promelas) exposed to a single nominal concentration of 17alpha-ethynylestradiol (EE2), a potent synthetic estrogen, under quasi-natural field conditions. Outdoor mesocosms were maintained under low, medium, and high nutrient supply conditions as categorized by total phosphorus (TP) level (nominal 0.012, 0.025, and 0.045 mg TP/L, respectively), and treated with EE2 with and without a carrier solvent. Using reverse transcription-polymerase chain reaction methods, vitellogenin gene (Vg) expression was determined in the fish collected at 0 h, 8 h, 24 h, 4 d, 7 d, and 14 d post-exposure. Induction of Vg was detected as early as 8h post-exposure, with and without the carrier solvent, and persisted through Day 14. Results showed Vg to be significantly greater at low nutrient levels (p<0.05), suggesting that EE2 bioavailability to the fish was likely greater under less-turbid water conditions.


Asunto(s)
Cyprinidae/metabolismo , Disruptores Endocrinos/toxicidad , Etinilestradiol/toxicidad , Eutrofización , Vitelogeninas/genética , Contaminantes Químicos del Agua/toxicidad , Animales , Ecosistema , Fertilizantes , Expresión Génica , Masculino , Nitrógeno/metabolismo , Fósforo/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pruebas de Toxicidad
20.
Reprod Toxicol ; 18(5): 687-700, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15219631

RESUMEN

We previously found that effects of perinatal dietary exposure to ethinylestradiol (EE) on the rat reproductive system differ depending on the diet used, with a more pronounced estrogenic impact with a regular diet that includes soy-derived proteins than with a soy-free (SF) diet. The present study was performed to examine whether genistein (GEN), a soy-derived major phytoestrogen, acts synergistically with EE. Maternal rats were fed SF diet without chemical (control) or containing 0.5-ppm EE, 0.5-ppm EE + 100-ppm GEN, 0.5-ppm EE + 1250-ppm GEN, or 1250-ppm GEN, from gestational day 15 to postnatal day (PND) 11. EE reduced serum testosterone in males at PND 3, and affected the onset of puberty of both sexes and estrous cyclicity and reproductive system in females, irrespective of co-administration of GEN. GEN alone also affected estrous cyclicity and the reproductive system in females. However, no combination effects of GEN with EE were evident, suggesting no synergism between the two.


Asunto(s)
Estrógenos/toxicidad , Etinilestradiol/toxicidad , Genisteína/administración & dosificación , Fitoestrógenos/administración & dosificación , Efectos Tardíos de la Exposición Prenatal , Reproducción/efectos de los fármacos , Administración Oral , Glándulas Suprarrenales/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Epidídimo/efectos de los fármacos , Estrógenos/administración & dosificación , Ciclo Estral/efectos de los fármacos , Etinilestradiol/administración & dosificación , Femenino , Genisteína/farmacología , Tamaño de la Camada/efectos de los fármacos , Masculino , Ovario/efectos de los fármacos , Fitoestrógenos/farmacología , Hipófisis/efectos de los fármacos , Hipófisis/patología , Embarazo , Ratas , Ratas Endogámicas , Diferenciación Sexual/efectos de los fármacos , Testículo/efectos de los fármacos , Testosterona/sangre , Útero/efectos de los fármacos , Útero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA