Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Intervalo de año de publicación
1.
Front Vet Sci ; 10: 1138564, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36992977

RESUMEN

Maternal nutrition during gestation has important effects on gene expression-mediated metabolic programming in offspring. To evaluate the effect of a protein-restricted maternal diet during gestation, pancreatic islets from male progeny of Wistar rats were studied at postnatal days (PND) 36 (juveniles) and 90 (young adults). The expression of key genes involved in ß-cell function and the DNA methylation pattern of the regulatory regions of two such genes, Pdx1 (pancreatic and duodenal homeobox 1) and MafA (musculoaponeurotic fibrosarcoma oncogene family, protein A), were investigated. Gene expression analysis in the pancreatic islets of restricted offspring showed significant differences compared with the control group at PND 36 (P < 0.05). The insulin 1 and 2 (Ins1 and Ins2), Glut2 (glucose transporter 2), Pdx1, MafA, and Atf2 (activating transcription factor 2), genes were upregulated, while glucokinase (Gck) and NeuroD1 (neuronal differentiation 1) were downregulated. Additionally, we studied whether the gene expression differences in Pdx1 and MafA between control and restricted offspring were associated with differential DNA methylation status in their regulatory regions. A decrease in the DNA methylation levels was found in the 5' flanking region between nucleotides -8118 to -7750 of the MafA regulatory region in restricted offspring compared with control pancreatic islets. In conclusion, low protein availability during gestation causes the upregulation of MafA gene expression in pancreatic ß-cells in the male juvenile offspring at least in part through DNA hypomethylation. This process may contribute to developmental dysregulation of ß-cell function and influence the long-term health of the offspring.

2.
Environ Toxicol ; 37(9): 2189-2200, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35596937

RESUMEN

Bisphenol S (BPS) has been introduced into the industry as a safer alternative to bisphenol A (BPA). However, the recent studies have reported a possible association between BPS and disturbed glucose homeostasis, indicating that it may be a risk factor for type 1 and type 2 diabetes mellitus, obesity, and gestational diabetes mellitus. Nevertheless, the role of BPS in glucose metabolism remains controversial. In this study, we investigated the glucose metabolism of male Wistar rats born from dams that were BPS-exposed (groups: BPS-L (0.05 mg/kg/day), BPS-H (20 mg/kg/day)) during pregnancy and lactation. We observed that both BPS treated groups of animals presented a significant decrease in anogenital distance/weight1/3 , as compared to control animals, although no alterations in testosterone levels were observed. Furthermore, the BPS-L group presented a significant decrease in body weight from postnatal day (PND) 21 to adult stage. In addition, a significant increase in glucose tolerance, pancreatic ß-cell proliferation, the frequency of small islets, and the average ß-cell size at PND 36 was observed in this group. However, no changes in insulin serum levels and percentage of ß-cells were recorded. Furthermore, these changes were not preserved at the adult stage (PND 120). The results suggest that the administration of low doses of BPS during the perinatal period induced an estrogenic like effect, with males apparently becoming more female-like in their responses to a glucose challenge.


Asunto(s)
Diabetes Mellitus Tipo 2 , Efectos Tardíos de la Exposición Prenatal , Animales , Compuestos de Bencidrilo/toxicidad , Femenino , Glucosa/metabolismo , Homeostasis , Humanos , Masculino , Exposición Materna/efectos adversos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Ratas Wistar
3.
Reprod Toxicol ; 86: 86-97, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31028817

RESUMEN

The aim was to evaluate the effect of perinatal BPA exposure of one or both parents on the implantation index and expression of talin, occludin and E-cadherin in the uterine epithelial cells (UEC) of the offspring. Pregnant Wistar dams (F0) received BPA or vehicle from gestational day (GD) 6 to lactation day 21. F1 animals were mated forming four groups: Control dam-Control sire (C♀-C♂), BPA dam -Control sire (B♀-C♂), Control dam -BPA sire (C♀-B♂), BPA dam -BPA sire (B♀-B♂). F1 dams were sacrificed at GD 6. Significantly decreased number of implantation sites was observed in the B♀-B♂ group as compared to the C♀-C♂ group, which correlated with decreased talin apical/basal expression ratio, occludin apical expression, and E-cadherin apical/lateral expression ratio in the UEC. Furthermore, decreased E-cadherin expression in the blastocyst was observed. Our data suggest that reduced protein expressions in F1 BPA offspring could result from decreased progesterone serum levels.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Cadherinas/metabolismo , Implantación del Embrión/efectos de los fármacos , Intercambio Materno-Fetal , Ocludina/metabolismo , Fenoles/toxicidad , Talina/metabolismo , Animales , Estradiol/sangre , Femenino , Masculino , Embarazo , Progesterona/sangre , Ratas Wistar , Útero/efectos de los fármacos , Útero/metabolismo
4.
J Steroid Biochem Mol Biol ; 178: 213-220, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29277708

RESUMEN

Parturient rats show a postpartum estrus, a period of sexual receptivity that occurs from 6 to 15 h after the birth of a litter, which allows the mother to gestate a second litter while simultaneously nursing the first one (lactating and pregnant). The present study investigated hormone levels and the expression pattern of estrogen receptor α, and ß, progesterone receptor isoforms and SRC1 in the hypothalamus and the preoptic area of lactating as well as in lactating-pregnant rats. In the latter, estradiol levels were 3-fold higher than those observed in lactating rats on day 14, meanwhile progesterone levels did not change in any condition. There were higher levels of prolactin in both lactating and lactating-pregnant rats on day 7 and decreased on the following days. In the hypothalamus of the lactating rat, the content of ERα increased during lactation meanwhile that of ERß decreased 50% on day 10. The content of both estrogen receptor subtypes in the hypothalamus increased 3-fold on day 21 in lactating-pregnant rats. In the preoptic area, the content of ERα was higher in lactating-pregnant rats on days 14 and 21 while the content of progesterone receptor isoforms was lower as compared with those found in lactating animals on days 7 and 10. The content of SRC1 increased 2-fold in the preoptic area only in lactating rats at day 14 and 21. These findings suggest that lactating- pregnant animals should exhibit differential neuroendocrine and molecular characteristics as compared to lactating animals.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Lactancia , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Animales , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley
5.
Reprod Toxicol ; 69: 106-120, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28216266

RESUMEN

We studied the effect of bisphenol-A (BPA) administration to rats, during the perinatal period, on the fertility of F1 generation and on the expression of tight junction (TJ) proteins in the uterus during early pregnancy. Pregnant Wistar dams (F0) received: BPA-L (0.05mg/kg/day), BPA-H (20mg/kg/day) or vehicle, from gestational day (GD) 6 to lactation day 21. F1 female pups were mated at 3 months of age and sacrificed at GD 1, 3, 6, and 7. Serum hormonal levels, ovulation rate, number of implantation sites and expression of TJ proteins in the uterus of F1 females were evaluated. BPA treatment induced no change in ovulation rate, but induced alterations in progesterone (P4) and estradiol (E2) serum levels, and in implantation rate. With regards to TJ proteins, BPA-H increased claudin-1 during all GDs; eliminated the peaks of claudins -3 and -4 at GD 3 and 6, respectively; and decreased claudin-7 at GD 6, ZO-1 from GD 1-6, and claudin-3 at GD 7 in stromal cells. BPA-L instead, eliminated claudin-3 peak at GD 3, increased claudin-4 and decreased claudin-7 from GD 1-6, decreased claudin-1 at GD 3 and 7 and claudin-4 at GD 7 in stromal cells. BPA-L also decreased ZO-1 at GDs 1 and 3 and increased ZO-1 at GD 6. Thus, BPA treatment during perinatal period perturbed, when the animals reached adulthood and became pregnant, the particular expression of TJ proteins in the uterine epithelium and reduced in consequence the number of implantation sites.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Proteínas de Uniones Estrechas/metabolismo , Útero/efectos de los fármacos , Animales , Implantación del Embrión/efectos de los fármacos , Estradiol/sangre , Femenino , Lactancia , Masculino , Embarazo , Progesterona/sangre , Ratas Wistar , Útero/metabolismo
6.
J Pharm Pharmacol ; 69(3): 305-313, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28134974

RESUMEN

OBJECTIVE: To design and synthesize an orcinol derivative compound, 3-formyl-2,4-dihydroxy-5,6-dimethyl sodium benzoate (A4), as an antioxidant molecule and to test its effects on oxidative stress in an in-vitro model of apoptosis of pancreatic rat beta cells induced by streptozotocin (STZ). METHODS: Scavenger properties of A4 were assessed using its capacity to capture the DPPH radical in vitro. Antiapoptotic properties of A4 were analysed by electron microscopy and TUNEL assay in rat pancreatic islets in a streptozotocin model. KEY FINDINGS: The results show that A4 displays antioxidant activity in in-vitro assays and induced a significant reduction in STZ-induced beta cell apoptosis and low ultrastructural damage to cellular organelles in the rat pancreatic islets as evidenced by electronic microscopy, this effect could be attributed to its antioxidant activity in a similar manner than resveratrol. CONCLUSION: The overall results indicate that the new orcinol derivative molecule displays both antioxidant and antiapoptotic effects and protect pancreatic beta cells against STZ damage.


Asunto(s)
Antioxidantes/farmacología , Islotes Pancreáticos/efectos de los fármacos , Resorcinoles/farmacología , Estreptozocina/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras , Ratas , Ratas Wistar
7.
Environ Toxicol ; 32(3): 856-868, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27240701

RESUMEN

Heptachlor has been targeted for global elimination because of its toxicity and environmental persistence, in accordance with the Stockholm Convention on Persistent Organic Pollutants (POPs). However, there is no regulation of heptachlor use in Mexico, where relatively high levels have been found in maternal breast milk. The aim of this study was to determine the effects of heptachlor on the reproductive system of offspring of two consecutive generations of rats (F1 and F2) from dams orally administered heptachlor during midgestation and lactation. Female offspring were analyzed for different phenotypic, reproductive, and molecular parameters. In the F1 generation, heptachlor treatment induced decreased body weight at weaning, increased female anogenital distance, and delayed vaginal opening. In both generations, serum progesterone levels decreased and estradiol levels remained unchanged, while overexpression of the progesterone receptor was observed in uterine epithelial cells on estrus day. In the F2 generation, expression of the estrogen receptor α increased in the glandular epithelium. Finally, heptachlor treatment did not affect apoptosis in the uterine epithelial cells. Overall, the results indicate that heptachlor induced female reproductive alterations when administered to dams during the perinatal period. Accordingly, exposure to heptachlor may represent a risk for the reproductive health of humans. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 856-868, 2017.


Asunto(s)
Heptacloro/toxicidad , Insecticidas/toxicidad , Reproducción/efectos de los fármacos , Animales , Cuello del Útero/metabolismo , Cuello del Útero/patología , Disruptores Endocrinos/toxicidad , Receptor alfa de Estrógeno/metabolismo , Femenino , Inmunohistoquímica , Lactancia , Masculino , Leche/química , Embarazo , Efectos Tardíos de la Exposición Prenatal , Progesterona/sangre , Pubertad/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Progesterona/metabolismo , Regulación hacia Arriba/efectos de los fármacos
8.
Oncol Lett ; 12(4): 2720-2727, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27703528

RESUMEN

Vascular endothelial growth factor (VEGF) is important in the growth and metastasis of cancer cells. In 2001, another angiogenic factor, endocrine gland-derived VEGF (EG-VEGF), was characterized and sequenced. EG-VEGF activity appears to be restricted to endothelial cells derived from endocrine glands. At the molecular level, its expression is regulated by hypoxia and steroid hormones. Although VEGF and EG-VEGF are structurally different, they function in a coordinated fashion. Since the majority of mammary tumors are hormone-dependent, it was hypothesized that EG-VEGF would be expressed in these tumors, and therefore, represent a potential target for anti-angiogenic therapy. The aim of the present study was to assess the expression of VEGF, EG-VEGF and its receptor (prokineticin receptor-1), as well as that of breast cancer resistant protein, estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, in 50 breast samples of infiltrating canalicular carcinoma (ICC) and their correlation with tumor staging. The samples were analyzed using reverse transcription-quantitative polymerase chain reaction and immunohistochemistry. Both angiogenic growth factors were identified in all samples. However, in 90% of the samples, the expression level of VEGF was significantly higher than that of EG-VEGF (P=0.024). There was no association between the expression of VEGF, EG-VEGF or its receptor with tumor stage. In ICC, the predominant angiogenic factor expressed was VEGF. The expression level of either factor was not correlated with the tumor-node-metastasis stage. Although ICC is derived from endothelial cells, EG-VEGF expression was not the predominant angiogenic/growth factor in ICC.

9.
J Endocrinol ; 231(1): 49-57, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27496224

RESUMEN

Maternal obesity (MO) is a deleterious condition that enhances susceptibility of adult offspring to metabolic diseases such as type 2 diabetes. The objective is to study the effect of MO on in vitro insulin secretion and pancreatic cellular population in offspring. We hypothesize that a harmful antenatal metabolic environment due to MO diminishes the basal glucose-responsive secretory function of pancreatic beta cells in offspring. Mothers were fed a control (C) or high-fat diet from weaning through pregnancy (120 days) and lactation. At postnatal days (PNDs) 36 and 110, pups were killed, peripheral blood was collected and pancreatic islets were isolated. Basal insulin secretion was measured in vitro in islets for 60 min. It was found that blood insulin, glucose and homeostasis model assessment (HOMA) index were unaffected by maternal diet and age in females. However, male MO offspring at PND 110 showed hyperinsulinemia and insulin resistance compared with C. Body weight was not modified by MO, but fat content was higher in MO pups compared with C pups. Triglycerides and leptin concentrations were higher in MO than in C offspring in all groups except in females at PND 36. Pancreatic islet cytoarchitecture was unaffected by MO. At PND 36, islets of male and female C and MO offspring responded similarly to glucose, but at PND 110, male and female MO offspring islets showed a 50% decrease in insulin secretion. It was concluded that MO impairs basal insulin secretion of offspring with a greater impact on males than females, and this effect mainly manifests in adulthood.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Obesidad/complicaciones , Efectos Tardíos de la Exposición Prenatal/sangre , Tejido Adiposo/metabolismo , Animales , Glucemia/análisis , Peso Corporal , Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa , Femenino , Insulina/sangre , Secreción de Insulina , Leptina/sangre , Masculino , Obesidad/sangre , Embarazo , Ratas , Triglicéridos/sangre
10.
Nutr Res ; 36(8): 855-62, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27440540

RESUMEN

A maternal low-protein (LP) diet programs fetal pancreatic islet ß-cell development and function and predisposes offspring to metabolic dysfunction later in life. We hypothesized that maternal protein restriction during pregnancy differentially alters ß- and α-cell populations in offspring by modifying islet ontogeny and function throughout life. We aimed to investigate the effect of an LP maternal diet on pancreatic islet morphology and cellular composition in female offspring on postnatal days (PNDs) 7, 14, 21, 36, and 110. Mothers were divided into 2 groups: during pregnancy, the control group (C) was fed a diet containing 20% casein, and the LP group was fed an isocaloric diet with 10% casein. Offspring pancreases were obtained at each PND and then processed. ß and α cells were detected by immunohistochemistry, and cellular area and islet size were quantified. Islet cytoarchitecture and total area were similar in C and LP offspring at all ages studied. At the early ages (PNDs 7-21), the proportion of ß cells was lower in LP than C offspring. The proportion of α cells was lower in LP than C offspring on PND 14 and higher on PND 21. The ß/α-cell ratio was lower in LP compared with C offspring on PNDs 7 and 21 and higher on PND 36 (being similar on PNDs 14 and 110). We concluded that maternal protein restriction during pregnancy modifies offspring islet cell ontogeny by altering the proportions of islet sizes and by reducing the number of ß cells postnatally, which may impact pancreatic function in adult life.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Páncreas/crecimiento & desarrollo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Femenino , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/crecimiento & desarrollo , Lactancia , Fenómenos Fisiologicos Nutricionales Maternos , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Wistar , Maduración Sexual , Destete
11.
Arch Med Res ; 46(2): 93-100, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25660337

RESUMEN

Nutrition during critical periods of development is one of the pivotal factors in establishing a lifelong healthy metabolism. Different nutritional deficiencies such as a low availability of proteins in the maternal diet produce alterations in offspring that include changes in insulin and glucose metabolism, a decrease in the size and number of cells of pancreatic islets of Langerhans, and premature ageing of the secretory function of pancreatic ß cells. Moreover, it has been reported that chronic nutritional stress is associated with epigenetic alterations in mechanisms of gene regulation during pancreatic development and function. These alterations can lead to dysfunctional states in pancreatic ß cells, which in the long run are responsible for the onset of metabolic diseases like type 2 diabetes. The present review summarizes the most important evidence in relation to the participation of epigenetic mechanisms in the regulation of gene expression during the intrauterine programming of the endocrine pancreas in animal models. Such mechanisms include DNA methylation as well as modifications of histones and microRNAs (miRNAs).


Asunto(s)
Epigénesis Genética/genética , Desarrollo Fetal/genética , Regulación del Desarrollo de la Expresión Génica/genética , Células Secretoras de Insulina/citología , Enfermedades Metabólicas/genética , Envejecimiento Prematuro/metabolismo , Animales , Metilación de ADN/genética , Diabetes Mellitus Tipo 2/genética , Dieta , Proteínas en la Dieta/farmacología , Femenino , Regulación de la Expresión Génica , Glucosa/metabolismo , Histonas/genética , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Desnutrición/metabolismo , MicroARNs/genética , Modelos Animales
12.
J Cereb Blood Flow Metab ; 34(6): 1001-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24667917

RESUMEN

Chronic consumption of high-fat-and-fructose diets (HFFD) is associated with the development of insulin resistance (InsRes) and obesity. Systemic insulin resistance resulting from long-term HFFD feeding has detrimental consequences on cognitive performance, neurogenesis, and long-term potentiation establishment, accompanied by neuronal alterations in the hippocampus. However, diet-induced hippocampal InsRes has not been reported. Therefore, we investigated whether short-term HFFD feeding produced hippocampal insulin signaling alterations associated with neuronal changes in the hippocampus. Rats were fed with a control diet or an HFFD consisting of 10% lard supplemented chow and 20% high-fructose syrup in the drinking water. Our results show that 7 days of HFFD feeding induce obesity and InsRes, associated with the following alterations in the hippocampus: (1) a decreased insulin signaling; (2) a decreased hippocampal weight; (3) a reduction in dendritic arborization in CA1 and microtubule-associated protein 2 (MAP-2) levels; (4) a decreased dendritic spine number in CA1 and synaptophysin content, along with an increase in tau phosphorylation; and finally, (5) an increase in reactive astrocyte associated with microglial changes. To our knowledge, this is the first report addressing hippocampal insulin signaling, as well as morphologic, structural, and functional modifications due to short-term HFFD feeding in the rat.


Asunto(s)
Astrocitos/metabolismo , Grasas de la Dieta/efectos adversos , Fructosa/efectos adversos , Hipocampo/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Neuritas/metabolismo , Transducción de Señal/efectos de los fármacos , Edulcorantes/efectos adversos , Sinapsis/metabolismo , Animales , Astrocitos/patología , Grasas de la Dieta/farmacocinética , Fructosa/farmacología , Hipocampo/patología , Masculino , Neuritas/patología , Ratas , Ratas Sprague-Dawley , Edulcorantes/farmacología , Sinapsis/patología
13.
Steroids ; 78(7): 717-25, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23583603

RESUMEN

We examined the ability of ICI 182,780 (ICI) to block uterine cell proliferation via protein kinase b/AKT pathway in the uterus of the rat during the estrous cycle. Intact rats, with regular estrous cycles, received a subcutaneous (s.c.) injection of either vehicle or ICI at 08:00 h on the day of proestrus or at 00:00 h on the day of estrus and sacrificed at 13:00 h of metaestrus. Estradiol (E2) and progesterone (P4) plasma levels were measured by radioimmunoassay. Both ICI treatments, induced a significant decrease (p<0.01) in uterine estrogen receptor alpha (ERα) content, had no effect on uterine progesterone receptor (PR) protein expression and caused marked nuclear localization of cyclin D1, in both luminal and glandular uterine epithelium, as compared to vehicle-treated animals. Furthermore, we detected that ICI treatment induced glycogen synthase kinase (Gsk3-ß) Ser 9 phosphorylation, which correlates with cyclin D1 nuclear localization. However, some differences were observed between the two different time schedules of administration. We observed that the administration of ICI at 08:00 h on proestrus day produced a 15% inhibition of luminal epithelial cell proliferation, reduced uterine wet weight by 21% and caused reduction of Akt phosphorylation at Ser 473 as compared to vehicle-treated animals, whereas ICI treatment at 00:00 h on estrus day had no effect on these parameters. The overall results indicate that ICI may exert agonistic and antagonistic effects on uterine cell proliferation through differential activation of the Akt pathway depending on the administration period during the estrous cycle, and indicates that the mechanism of cell proliferation during the physiological conditions of the estrous cycle, is under a different and more complex regulation than in the ovariectomized + E2 animal model.


Asunto(s)
Estradiol/análogos & derivados , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Útero/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Femenino , Fulvestrant , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Inmunohistoquímica , Proteínas Proto-Oncogénicas c-akt/genética , Radioinmunoensayo , Ratas , Útero/efectos de los fármacos
14.
Curr Top Med Chem ; 11(13): 1728-35, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21463250

RESUMEN

Traditionally the role of sexual steroid hormones was focused primarily on reproductive organs: the breast, female reproductive tract (uterus, mammary gland, and ovary), and male reproductive tract (testes, epididymis and prostate), however our current understanding of tissue-specific effects of sex steroids has elucidated new aspects in its functionality. Recent data have shown that many other tissues are targets of those hormones in addition to classical reproductive organs. The pancreas (which performs both endocrine and exocrine functions), has proven to be an extragonadal target of sexual steroid hormone action. The endocrine pancreas has a pivotal role on carbohydrate homeostasis and deterioration in function produces diabetes. Diabetes is a metabolic disorder that has high prevalence worldwide, particularly in developing countries. It has been shown that steroid hormones have an important role in susceptibility and development of diabetes in animal models, in humans its role is less clear, however the most evident effect is on the perimenopausal women, in this stage the decrease in gonadal steroids produces an increase on susceptibility to develop diabetes mellitus; in men, hypoandrogenism is associated with an increased prevalence of insulin resistance. This review focused on the effects of sexual steroids on pancreatic function and diabetes.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Hormonas Esteroides Gonadales/farmacología , Islotes Pancreáticos/efectos de los fármacos , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Femenino , Humanos , Masculino
15.
Horm Behav ; 59(1): 37-43, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20950620

RESUMEN

We studied the role of progesterone receptor (PR) in the display of female sexual behavior during postpartum estrus in rats. Adult female rats were treated with the PR antagonist, RU486 (1.25 and 5 mg), 3 h after parturition and sexual behavior was evaluated throughout the first postpartum day. Estradiol and progesterone serum levels changed during the first 24 h postpartum. The highest estradiol and progesterone levels were found at 9 and 12 h postpartum, respectively. The predominant PR isoform in the hypothalamus and the preoptic area was PR-A during postpartum day. The content of PR-A increased at 6 h postpartum in the hypothalamus and the preoptic area, and decreased in both regions at 9 h. PR-B content only increased in the preoptic area at 12 h postpartum. The highest display of lordotic and proceptive behaviors were found at 12 h postpartum. The treatment with 1.25 and 5 mg of RU486 respectively reduced lordosis by 61% and 92% at 12 h postpartum. These results suggest that PR is essential in the display of postpartum estrus in rats.


Asunto(s)
Estro/metabolismo , Hipotálamo/metabolismo , Periodo Posparto/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/metabolismo , Conducta Sexual Animal/fisiología , Análisis de Varianza , Animales , Western Blotting , Estradiol/sangre , Estro/efectos de los fármacos , Femenino , Hipotálamo/efectos de los fármacos , Mifepristona/farmacología , Periodo Posparto/efectos de los fármacos , Postura/fisiología , Progesterona/sangre , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Conducta Sexual Animal/efectos de los fármacos , Factores de Tiempo
16.
Reprod Toxicol ; 31(2): 177-83, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21055461

RESUMEN

Bisphenol A (BPA) is an estrogenic agonist compound that induces changes in diverse reproductive parameters in rats. The aim of the present study was to determine the effects of BPA given in drinking water containing 10mg/L (approximate dose 1.2mg/kg BW/day), administered chronically to rats during pregnancy and lactation, on reproductive tract parameters of the offspring. 79.2% of the female offspring from BPA-treated mothers presented irregular estrous cycles. As compared to the control group, a significant increase in the thickness of the uterine epithelia and stroma was observed in the BPA group. Additionally, 60% of the female offspring from BPA mothers did not undergo abundant uterine epithelial apoptosis during the estrus phase of the cycle while control animals did. In addition, a down regulation of ERα expression was observed in epithelial cells on estrus day. The results indicate that BPA, when administered chronically in water beverages to dams, modifies the reproductive cycle of the offspring during young adulthood.


Asunto(s)
Estrógenos no Esteroides/toxicidad , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal , Reproducción/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Compuestos de Bencidrilo , Epitelio/anatomía & histología , Epitelio/química , Epitelio/efectos de los fármacos , Receptor alfa de Estrógeno/análisis , Estrógenos no Esteroides/administración & dosificación , Ciclo Estral/efectos de los fármacos , Femenino , Lactancia , Fenoles/administración & dosificación , Embarazo , Ratas , Ratas Wistar , Reproducción/fisiología , Útero/anatomía & histología , Útero/química , Útero/efectos de los fármacos , Agua
17.
J Steroid Biochem Mol Biol ; 122(4): 107-13, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20580673

RESUMEN

The endocrine pancreas is central in the physiopathology of diabetes mellitus. Nutrients and hormones control endocrine pancreatic function and the secretion of insulin and other pancreatic islet hormones. Although the pancreas is not usually considered as a target of steroids, increasing evidence indicates that sex steroid hormones modify pancreatic islet function. The biological effects of steroid hormones are transduced by both, classical and non-classical steroid receptors that in turn produce slow genomic and rapid non-genomic responses. In this review, we focused on the effects of sex steroid hormones on endocrine pancreatic function, with special emphasis in animal studies.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus/metabolismo , Humanos
18.
Pancreas ; 39(2): 193-200, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20093993

RESUMEN

OBJECTIVES: To investigate the protective role of steroid hormones on streptozotocin (STZ)-induced apoptosis in rat pancreatic beta cells. METHODS: Two sets of experiments were performed. In the first, male rats were orchidectomized and substituted 72 hours later with testosterone, estradiol, or progesterone, and 24 hours later, administered with STZ. Subjects were killed 6 hours later, and apoptosis was determined in sections of the pancreas. In the second experiment, male or female rats were gonadectomized, were further substituted with testosterone, and then administered STZ. Six hours later, the animals were killed, and apoptosis, as well as immunoreactive expression of insulin, catalase, or Cu/Zn superoxide dismutase, was determined in sections of the pancreas. In addition, gonadectomized male or female subjects were substituted with testosterone and administered STZ, and 24 hours later, serum glucose and insulin were measured. RESULTS: It was found that the cytoprotective effect was only shown in testosterone-treated male rats but not progesterone- or estradiol-treated male rats. In addition, the effect was seen in male rats but not in female rats, and there was an inverse correlation between apoptotic index and antioxidant enzyme immunoreactivity. CONCLUSIONS: The cytoprotective effect of testosterone is sex specific and is related to the induction of antioxidant enzyme activities in pancreatic beta cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/prevención & control , Células Secretoras de Insulina/metabolismo , Testosterona/metabolismo , Animales , Glucemia/metabolismo , Catalasa/metabolismo , Citoprotección , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Estradiol/metabolismo , Femenino , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Insulina/sangre , Células Secretoras de Insulina/patología , Masculino , Orquiectomía , Ovariectomía , Progesterona/metabolismo , Ratas , Ratas Wistar , Factores Sexuales , Estreptozocina , Superóxido Dismutasa/metabolismo
19.
J Steroid Biochem Mol Biol ; 113(3-5): 259-68, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19429431

RESUMEN

The rodent uterus is a widely studied target tissue for sexual steroid hormone action. The aim of the present study was to assess the molecular mechanism that participates in the initiation of cell proliferation of the rat uterine epithelial cells during the estrus (E)-metestrus (M) transition. Cell proliferation, ERalpha, c-fos, cyclin D1 and D3, cdk4, and cdk6 proteins were assessed in these animals by immunohistochemistry. Estradiol (E(2)) and progesterone (P(4)) plasma levels were assessed by RIA. The results indicate that the glandular epithelium starts to proliferate at 21:00 h on estrus day, and initiates at least 3h before the luminal epithelium does. Fos expression was markedly increased during the afternoon of estrus day, and its increase was in parallel to ERalpha expression. Interestingly, both, cyclin D1 and D3 were abundantly expressed in the luminal and glandular epithelia, and nuclear immunolabelling of cyclin D1 and D3 precedes BrdU incorporation in the cell. cdk4 and cdk6 were localized in the nuclei in both epithelia throughout the studied time course. In addition, cdk4 was more abundant throughout estrus and metestrus days than cdk6. The overall results indicate that ERalpha, Fos and cyclins D1 and D3, cdk4 and cdk6 are expressed in both glandular and luminal epithelia of the rat uterus during the E-M transition. In conclusion, there is a good correlation between sequential expression of these proteins and cell cycle progression in the rat uterine epithelial cells during the estrous cycle. However, the differences observed in the cellular localization, time course of expression and the cellular types that express both cyclins between physiological and pharmacological conditions, demonstrated different mechanisms of regulation and should be due to the complex hormonal milieu during the estrous cycle.


Asunto(s)
Proliferación Celular , Células Epiteliales/fisiología , Ciclo Estral/fisiología , Útero/citología , Animales , Ciclo Celular/fisiología , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D3 , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Células Epiteliales/citología , Estradiol/sangre , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Proteínas Oncogénicas v-fos/genética , Proteínas Oncogénicas v-fos/metabolismo , Progesterona/sangre , Ratas , Ratas Wistar , Útero/fisiología
20.
Mol Reprod Dev ; 76(6): 564-72, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19117026

RESUMEN

Estrous cycle in mammals includes marked epithelial changes in reproductive tract, regulated by sex steroid hormones. In the present work we studied the activation of caspases and apoptotic pattern in uterine epithelial cells during proestrus and estrus, and the effect of mating in this process. In addition, we investigated the role of seminal vesicle secretions on apoptosis of uterine epithelia. Apoptotic index was evaluated by TUNEL assay, caspases-8, -9, and -3 activation was detected by Western blot and active caspase-3 expression was detected by immunohistochemistry. Our results show that mating during proestrus and estrus transition induced changes in the apoptotic pattern of uterine luminal epithelium during estrus, characterized by a delay in the onset of apoptosis as compared with that observed in nonmated rats. No differences in the apoptotic pattern in the glandular epithelium between mated and nonmated rats were observed. Seminal vesicle secretions inhibited luminal epithelium apoptosis, while no changes in glandular epithelium apoptosis were observed. We also demonstrate that activation of caspases-8, -9, and -3 occurred in both mated and nonmated rats. Active caspase-3 was detected in the luminal and glandular epithelium in both nonmated and mated rats. The overall results indicate that mating delays but does not prevent the cellular death of the rat uterine luminal epithelium and seminal vesicle secretions are involved in this delay. Finally, the activation of both the mitochondrial and the membrane receptor pathways of cell death are implicated in the molecular mechanism of uterine apoptosis.


Asunto(s)
Apoptosis/fisiología , Células Epiteliales/fisiología , Ciclo Estral/fisiología , Conducta Sexual Animal/fisiología , Útero/citología , Animales , Caspasas/metabolismo , Activación Enzimática , Células Epiteliales/citología , Estradiol/sangre , Femenino , Masculino , Progesterona/sangre , Ratas , Ratas Wistar , Vesículas Seminales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...