Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Endocrinol ; 261(3)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38579817

RESUMEN

Exposure to glyphosate-based herbicides (GBH) and consumption of cafeteria (CAF) diet, which are widespread in Western society, seem to be associated with endometrial hyperplasia (EH). Here, we aimed to evaluate the effects of a subchronic low dose of GBH added to the CAF diet on the rat uterus. Female Wistar rats were fed from postnatal day (PND)21 until PND240 with chow (control) or CAF diet. Since PND140, rats also received GBH (2 mg of glyphosate/kg/day) or water through food, yielding four experimental groups: control, CAF, GBH, and CAF+GBH. On PND240, CAF and CAF+GBH animals showed an increased adiposity index. With respect to the control group, no changes in the serum levels of 17ß-estradiol and progesterone were found. However, progesterone levels were higher in the CAF+GBH group than in the CAF and GBH groups. In the uterus, both studied factors alone and in combination induced morphological and molecular changes associated with EH. Furthermore, the addition of GBH provoked an increased thickness of subepithelial stroma in rats fed with the CAF diet. As a consequence of GBH exposure, CAF+GBH rats exhibited an increased density of abnormal gland area, considered preneoplastic lesions, as well as a reduced PTEN and p27 expression, both tumor suppressor molecules that inhibit cell proliferation, with respect to control rats. These results indicate that the addition of GBH exacerbates the CAF effects on uterine lesions and that the PTEN/p27 signaling pathway seems to be involved. Further studies focusing on the interaction between unhealthy diets and environmental chemicals should be encouraged to better understand uterine pathologies.


Asunto(s)
Glicina , Glifosato , Herbicidas , Ratas Wistar , Útero , Animales , Femenino , Útero/efectos de los fármacos , Útero/patología , Útero/metabolismo , Herbicidas/toxicidad , Glicina/análogos & derivados , Ratas , Hiperplasia Endometrial/inducido químicamente , Hiperplasia Endometrial/patología , Hiperplasia Endometrial/metabolismo , Progesterona/sangre , Dieta , Estradiol/sangre , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética
2.
Mol Cell Endocrinol ; 585: 112198, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38467370

RESUMEN

The aim of this study was to investigate uterine lesions, uterine endocrine status and expression of genes involved in uterine differentiation in a rat model of polycystic ovary syndrome (PCOS). The possible involvement of the androgen receptor (AR) was also investigated. PCOS rats showed an increased incidence of uterine epithelial and glandular lesions and elevated serum testosterone level, which was not detected in uterine tissue. Uterine 17ß-estradiol, estrone and progesterone were detected in 100%, 75% and 50% of the animals, respectively. This was associated with a decrease in Star and an increase in Hsd17b2, Srd5a1 and Cyp19a1, suggesting that uterine steroids are not synthesized de novo in PCOS and that alterations in these enzymes may explain the absence of testosterone and low progesterone. In addition, ESR2 decreased and AR increased, suggesting possible steroid receptor crosstalk. Genes associated with uterine differentiation, PTEN and WNT5a, also showed reduced expression. PCOS rats treated with flutamide, an AR antagonist, were similar to PCOS rats in terms of uterine lesions, serum steroid levels, ESR2, PTEN and WNT5a expression. However, testosterone, AR and aromatase levels were similar to control rats, with decreased expression of ESR1 and HOXA10, suggesting that these expressions are AR dependent. Our results suggest that the primary cause of the observed uterine lesions in the PCOS rat model is the altered endocrine status and consequently changes in genes related to uterine differentiation.


Asunto(s)
Síndrome del Ovario Poliquístico , Femenino , Humanos , Ratas , Animales , Síndrome del Ovario Poliquístico/metabolismo , Progesterona , Estradiol , Testosterona , Esteroides
3.
J Nutr Biochem ; 122: 109451, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37748623

RESUMEN

Mesolimbic dopaminergic circuit is essential for food reward and motivational behaviors and can contribute to weight gain and obesity. Litter reduction is a classical model for studying the effects of neonatal overfeeding and overweight. Litters of Wistar rats were reduced to 4 pups/dam for small litter (SL) and 10 pups/dam for normal litter at postnatal day (PND) 4. Immediately after performing the feeding behavior tests, the animals were sacrificed in PND21 and PND90. The ventral tegmental area (VTA), Nucleus Accumbens Core (NAcC) and Shell (NAcSh) were isolated from frozen brain sections using the Palkovits micropunch technique. RNA and DNA were extracted from these areas, gene expression was measured by RT-qPCR and DNA methylation levels were measured by MSRM-qPCR technique. SL-PND21 animals presented increased expression levels of Tyrosine Hydroxylase and Dopamine Receptor D2 in VTA, decreased expression levels of dopamine active transporter (DAT) in VTA, and higher expression levels of DAT in NAcC. On the other hand, SL-PND90 animals showed decreased expression levels of Dopamine Receptor D1 and higher expression of DAT in NAcSh. These animals also evidenced impaired sensory-specific satiety. In addition, altered promoter methylation was observed at weaning, and remained in adulthood. This work demonstrates that neonatal overfeeding induces disruptions in the mesolimbic dopaminergic circuitry and causes alterations in feeding behavior from weaning to adulthood, suggesting that the neonatal period is critical for the normal development of dopaminergic circuit that impact on feeding behavior.


Asunto(s)
Metilación de ADN , Dopamina , Ratas , Animales , Dopamina/metabolismo , Ratas Wistar , Conducta Alimentaria , Núcleo Accumbens/metabolismo
4.
J Nutr Biochem ; 106: 109015, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35447318

RESUMEN

Maternal diet has impact on reproduction, fetal development and offspring behavior, although molecular mechanisms remained unknown. Our aims were to assess (1) the effects of a cafeteria (CAF) diet (western diet habits) on female reproductive performance, fetal and placental parameters on gestational day 21 and litter size and pup weight at birth; and (2) placental messenger RNA (mRNA) expression and epigenetic regulation of Insulin-Like Growth Factor (Igf) and Vascular Endothelial Growth Factor (Vegf) and their receptors. Female Wistar rats were fed with control or CAF diet from weaning until parturition. At week 14 after diets started, females were mated and half of the animals were euthanized on gestational day 21 to evaluate reproductive parameters including the pregnancy rate, number of corpora lutea, implantation sites and resorption sites. Moreover, fetal weight and length, placental weight, and placental index were recorded. Placentas were collected for mRNA quantification and DNA methylation analysis. The remaining animals were allowed to give birth and the number and weight of the pups were evaluated. CAF diet did not affect reproductive performance or fetal weight and length. However, CAF-fed animals showed a decrease in placental weight and index and the pups exhibited a low birth weight. Additionally, we found an upregulation of Igf2 and a down regulation of Vegf placental mRNA expression in CAF dams, associated with methylation status changes of their promoters. We conclude that female chronic CAF diet consumption impairs feto-placental development and could be explained by an epigenetic disruption of Igf and Vegf systems.


Asunto(s)
Placenta , Factor A de Crecimiento Endotelial Vascular , Animales , Dieta , Epigénesis Genética , Femenino , Peso Fetal , Placenta/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Vitam Horm ; 118: 171-198, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35180926

RESUMEN

Neurosteroids are steroids synthesized de novo from cholesterol in brain regions, and regulate processes associated with the development and functioning of the nervous system. Enzymes and proteins involved in the synthesis of these steroids have been detected in several brain regions, including hippocampus, hypothalamus, and cerebral cortex. Hippocampus has long been associated with learning and memory functions, while the loss of its functionality has been linked to neurodegenerative pathologies. In this sense, neurosteroids are critical for the maintenance of hippocampal functions and neuroprotective effects. Moreover, several factors have been shown to deregulate expression of steroidogenic enzymes in the rodent brain, including aging, enrichment experiences, diet habits, drug/alcohol consumption, hormone fluctuations, neurodegenerative processes and other diseases. These transcriptional deregulations are mediated mainly by transcription factors and epigenetic mechanisms. An epigenetic modification of chromatin involves changes in bases and associated proteins in the absence of changes in the DNA sequence. One of the most well-studied mechanisms related to gene silencing is DNA methylation, which involves a reversible addition of methyl groups in a cytosine base. Importantly, these epigenetic marks could be maintained over time and could be transmitted transgenerationally. The aim of this chapter is to present the most relevant steroidogenic enzymes described in rodent hippocampus; to discuss about their transcriptional regulation under different conditions; to show the main gene control regions and to propose DNA methylation as an epigenetic mechanism through which the expression of these enzymes could be controlled.


Asunto(s)
Regulación de la Expresión Génica , Hipocampo , Metilación de ADN , Epigénesis Genética , Hipocampo/metabolismo
7.
Front Endocrinol (Lausanne) ; 12: 672532, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305812

RESUMEN

Glyphosate base herbicides (GBHs) are the most widely applied pesticides in the world and are mainly used in association with GBH-tolerant crop varieties. Indiscriminate and negligent use of GBHs has promoted the emergence of glyphosate resistant weeds, and consequently the rise in the use of these herbicides. Glyphosate, the active ingredient of all GBHs, is combined with other chemicals known as co-formulants that enhance the herbicide action. Nowadays, the safety of glyphosate and its formulations remain to be a controversial issue, as evidence is not conclusive whether the adverse effects are caused by GBH or glyphosate, and little is known about the contribution of co-formulants to the toxicity of herbicides. Currently, alarmingly increased levels of glyphosate have been detected in different environmental matrixes and in foodstuff, becoming an issue of social concern. Some in vitro and in vivo studies have shown that glyphosate and its formulations exhibit estrogen-like properties, and growing evidence has indicated they may disrupt normal endocrine function, with adverse consequences for reproductive health. Moreover, multigenerational effects have been reported and epigenetic mechanisms have been proved to be involved in the alterations induced by the herbicide. In this review, we provide an overview of: i) the routes and levels of human exposure to GBHs, ii) the potential estrogenic effects of glyphosate and GBHs in cell culture and animal models, iii) their long-term effects on female fertility and mechanisms of action, and iv) the consequences on health of successive generations.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Glicina/análogos & derivados , Herbicidas/toxicidad , Infertilidad Femenina/inducido químicamente , Reproducción/efectos de los fármacos , Femenino , Glicina/toxicidad , Humanos , Glifosato
8.
Front Endocrinol (Lausanne) ; 12: 671991, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093442

RESUMEN

Glyphosate is a phosphonomethyl amino acid derivative present in a number of non-selective and systemic herbicides. During the last years the use of glyphosate-based herbicide (GBH) has been increasing exponentially around the world, including Argentina. This fact added to the detection of glyphosate, and its main metabolite, amino methylphosphonic acid (AMPA), in environmental matrices such as soil, sediments, and food, has generated great concern about its risks for humans, animals, and environment. During the last years, there were controversy and intense debate regarding the toxicological effects of these compounds associated with the endocrine system, cancer, reproduction, and development. The mechanisms of action of GBH and their metabolites are still under investigation, although recent findings have shown that they could comprise epigenetic modifications. These are reversible mechanisms linked to tissue-specific silencing of gene expression, genomic imprinting, and tumor growth. Particularly, glyphosate, GBH, and AMPA have been reported to produce changes in global DNA methylation, methylation of specific genes, histone modification, and differential expression of non-coding RNAs in human cells and rodents. Importantly, the epigenome could be heritable and could lead to disease long after the exposure has ended. This mini-review summarizes the epigenetic changes produced by glyphosate, GBHs, and AMPA in humans and rodents and proposes it as a potential mechanism of action through which these chemical compounds could alter body functions.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Glicina/análogos & derivados , Herbicidas/toxicidad , Reproducción/efectos de los fármacos , Animales , Metilación de ADN/efectos de los fármacos , Glicina/toxicidad , Mamíferos , Glifosato
9.
Mol Cell Endocrinol ; 522: 111123, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33338550

RESUMEN

A small litter (SL) model was used to determine how neonatal overfeeding affects the homeostatic control of food intake in male rats at weaning and postnatal day (PND) 90. At PND4, litters were reduced to small (4 pups/dam) or normal (10 pups/dam) litters. At weaning, SL rats showed higher body weight and characteristic features of the metabolic syndrome. Gene expression of pro-opiomelanocortin (POMC), cocaine and amphetamine regulated transcript, neuropeptide Y (NPY) and leptin and ghrelin (GHSR) receptors were increased and POMC promoter was hypomethylated in arcuate nucleus, indicating that the early development of obesity may involve the GHSR/NPY system and changes in POMC methylation state. At PND90, body weight, metabolic parameters and gene expression were restored; however, POMC methylation state remained altered. This work provides insight into the effects of neonatal overfeeding, showing the importance of developmental plasticity in restoring early changes in central pathways involved in metabolic programming.


Asunto(s)
Metilación de ADN/genética , Conducta Alimentaria , Homeostasis , Proopiomelanocortina/genética , Regiones Promotoras Genéticas , Tejido Adiposo/metabolismo , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/metabolismo , Peso Corporal/genética , Islas de CpG/genética , Epidídimo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Masculino , Metaboloma , Neuropéptidos/genética , Neuropéptidos/metabolismo , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Factores de Tiempo
10.
Mol Cell Endocrinol ; 485: 44-53, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30721712

RESUMEN

In the present study, we analyzed the effects of a short-term environmental enrichment on the mRNA expression and DNA methylation of steroidogenic enzymes in the hippocampus. Thus, young adult (80-day-old) and middle-aged (350-day-old) Wistar female rats were exposed to sensory (SE) or motor (ME) enrichment during 10 days and compared to animals housed under standard conditions. SE was provided by an assortment of objects that included plastic tubes and toys; for ME, rodent wheels were provided. In young adult animals, SE and ME increased the mRNA expression of cytochrome P450 17α-hydroxylase/c17,20-lyase, steroid 5α-reductase type 1 (5αR-1) and 3α-hydroxysteroid dehydrogenase and decreased the methylation levels of 5αR-1 gene. In middle-aged rats, ME and SE upregulated the gene expression of aldosterone synthase and decreased the methylation state of its promoter. These results propose that SE and ME differentially regulate the transcription of neurosteroidogenic enzymes through epigenetic mechanisms in young and aged rats.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/genética , Citocromo P-450 CYP11B2/genética , Metilación de ADN , Esteroide 17-alfa-Hidroxilasa/genética , Regulación hacia Arriba , Animales , Vías Biosintéticas , Epigénesis Genética , Femenino , Hipocampo , Vivienda para Animales , Estimulación Luminosa , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Carrera
11.
Mol Cell Endocrinol ; 450: 113-125, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28479374

RESUMEN

We evaluated the effect of cafeteria diet (CAF) on the mRNA levels and DNA methylation state of feeding-related neuropeptides, and neurosteroidogenic enzymes in discrete hypothalamic nuclei. Besides, the expression of steroid hormone receptors was analyzed. Female rats fed with CAF from weaning increased their energy intake, body weight, and fat depots, but did not develop metabolic syndrome. The increase in energy intake was related to an orexigenic signal of paraventricular (PVN) and ventromedial (VMN) nuclei, given principally by upregulation of AgRP and NPY. This was mildly counteracted by the arcuate nucleus, with decreased AgRP expression and increased POMC and kisspeptin expression. CAF altered the transcription of neurosteroidogenic enzymes in PVN and VMN, and epigenetic mechanisms associated with differential promoter methylation were involved. The changes observed in the hypothalamic nuclei studied could add information about their differential role in food intake control and how their action is disrupted in obesity.


Asunto(s)
Metilación de ADN/genética , Dieta , Ingestión de Alimentos/genética , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Tejido Adiposo/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Sitios de Unión , Peso Corporal , Simulación por Computador , Ingestión de Energía/genética , Femenino , Prueba de Tolerancia a la Glucosa , Neuropéptidos/genética , Neuropéptidos/metabolismo , Tamaño de los Órganos/genética , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Esteroides/metabolismo , Transcripción Genética , Núcleo Hipotalámico Ventromedial/metabolismo
12.
Mol Cell Endocrinol ; 401: 111-9, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25486512

RESUMEN

The absence of phytoestrogens in the diet during pregnancy has been reported to result in obesity later in adulthood. We investigated whether phytoestrogen withdrawal in adult life could alter the hypothalamic signals that regulate food intake and affect body weight and glucose homeostasis. Male Wistar rats fed from conception to adulthood with a high phytoestrogen diet were submitted to phytoestrogen withdrawal by feeding a low phytoestrogen diet, or a high phytoestrogen-high fat diet. Withdrawal of dietary phytoestrogens increased body weight, adiposity and energy intake through an orexigenic hypothalamic response characterized by upregulation of AGRP and downregulation of POMC. This was associated with elevated leptin and T4, reduced TSH, testosterone and estradiol, and diminished hypothalamic ERα expression, concomitant with alterations in glucose tolerance. Removing dietary phytoestrogens caused manifestations of obesity and diabetes that were more pronounced than those induced by the high phytoestrogen-high fat diet intake.


Asunto(s)
Glucemia/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/metabolismo , Obesidad/etiología , Fitoestrógenos/administración & dosificación , Alimentación Animal , Animales , Peso Corporal/efectos de los fármacos , Suplementos Dietéticos , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Masculino , Fitoestrógenos/farmacología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA