Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Ovarian Res ; 16(1): 138, 2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37443082

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder that frequently exhibits low-grade inflammation, pro-oxidant activity, and gut dysbiosis. PCOS has become one of the leading causes of female infertility worldwide. Recently, omega-3 polyunsaturated fatty acids (PUFAs) have been proven to benefit metabolic disorders in PCOS patients. However, its roles in the regulation of metabolic and endocrinal balances in PCOS pathophysiology are not clear. In the present study, we aimed to explore how omega-3 PUFAs alleviate ovarian dysfunction and insulin resistance in mice with dehydroepiandrosterone (DHEA)-induced PCOS by modulating the gut microbiota. METHODS: We induced PCOS in female mice by injecting them with DHEA and then treated them with omega-3 PUFAs. 16S ribosomal DNA (rDNA) amplicon sequencing, fecal microbiota transplantation (FMT) and antibiotic treatment were used to evaluate the role of microbiota in the regulation of ovarian functions and insulin resistance (IR) by omega-3 PUFAs. To further investigate the mechanism of gut microbiota on omega-3-mediated ovarian and metabolic protective effects, inflammatory and oxidative stress markers in ovaries and thermogenic markers in subcutaneous and brown adipose tissues were investigated. RESULTS: We found that oral supplementation with omega-3 PUFAs ameliorates the PCOS phenotype. 16S rDNA analysis revealed that omega-3 PUFA treatment increased the abundance of beneficial bacteria in the gut, thereby alleviating DHEA-induced gut dysbiosis. Antibiotic treatment and FMT experiments further demonstrated that the mechanisms underlying omega-3 benefits likely involve direct effects on the ovary to inhibit inflammatory cytokines such as IL-1ß, TNF-α and IL-18. In addition, the gut microbiota played a key role in the improvement of adipose tissue morphology and function by decreasing multilocular cells and thermogenic markers such as Ucp1, Pgc1a, Cited and Cox8b within the subcutaneous adipose tissues. CONCLUSION: These findings indicate that omega-3 PUFAs ameliorate androgen-induced gut microbiota dysbiosis. The gut microbiota plays a key role in the regulation of omega-3-mediated IR protective effects in polycystic ovary syndrome mice. Moreover, omega-3 PUFA-regulated improvements in the ovarian dysfunction associated with PCOS likely involve direct effects on the ovary to inhibit inflammation. Our findings suggest that omega-3 supplementation may be a promising therapeutic approach for the treatment of PCOS by modulating gut microbiota and alleviating ovarian dysfunction and insulin resistance.


Asunto(s)
Suplementos Dietéticos , Ácidos Grasos Omega-3 , Microbioma Gastrointestinal , Síndrome del Ovario Poliquístico , Animales , Femenino , Ratones , Deshidroepiandrosterona/toxicidad , Microbioma Gastrointestinal/fisiología , Resistencia a la Insulina , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/fisiopatología , Ácidos Grasos Omega-3/uso terapéutico
2.
J Ovarian Res ; 15(1): 11, 2022 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-35057828

RESUMEN

BACKGROUND: Melatonin, as a free radical scavenger exhibiting genomic actions, regulates the antioxidant genes expression and apoptosis mechanisms. In polycystic ovary syndrome (PCOS) patients, an imbalance between free radicals and antioxidants in follicular fluid leads to oxidative stress, aberrant folliculogenesis, and intrinsic defects in PCOS oocytes. In this experimental mouse model study, oocytes of PCOS and the control groups were cultured in different melatonin concentrations (10- 5, 10- 6, and 10- 7 M) to investigate the expression of oocyte maturation-related genes (Gdf9/Bmp15), antioxidant-related genes (Gpx1/Sod1), apoptotic biomarkers (Bcl2/Bax) and total intracellular ROS levels. RESULTS: Gdf9 and Bmp15, Gpx1 and Sod1 were up-regulated in PCOS and control oocytes cultured in all melatonin concentrations compared to those cultured in IVM basal medium (P < 0.05). A significant decrease in the total ROS level was observed in all groups cultured in the supplemented cultures. Melatonin increased Bcl2 and decreased Bax gene expression in PCOS and control oocytes compared to non-treated oocytes. CONCLUSIONS: Melatonin increased antioxidant gene expression and regulated the apoptosis pathway, effectively reducing the adverse effects of culture conditions on PCOS oocytes. Furthermore, it influenced the expression of oocyte maturation-related genes in PCOS, providing valuable support during the IVM process.


Asunto(s)
Antioxidantes/metabolismo , Melatonina/farmacología , Oocitos/efectos de los fármacos , Oogénesis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína Morfogenética Ósea 15/genética , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Femenino , Glutatión Peroxidasa/genética , Factor 9 de Diferenciación de Crecimiento/genética , Técnicas de Maduración In Vitro de los Oocitos , Ratones , Oocitos/metabolismo , Oogénesis/genética , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa-1/genética , Proteína X Asociada a bcl-2/genética , Glutatión Peroxidasa GPX1
3.
Peptides ; 145: 170624, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34375684

RESUMEN

OBJECTIVE: The purpose of this study was to explore the effect of dulaglutide on DHEA induced PCOS rats and its mechanism, to provide new drugs and research directions for clinical treatment of PCOS. METHODS: In this study, the PCOS model was established by giving female SD rats subcutaneous injection of DHEA for 21 consecutive days. After modeling, the treatment group was injected subcutaneously with three doses of dulaglutide for 3 weeks. The model group was injected with sterile ultrapure water, and the normal group did not get any intervention. The body weight changes of rats in each group were recorded from the first day when rats received the administration of dulaglutide. Three weeks later, the rats were fasted the night after the last treatment, determined fasting insulin and fasting glucose the next day. After the rats were anesthetized by chloral hydrate, more blood was collected from the heart of the rat. The serum insulin, testosterone and sex hormone binding globulin (SHBG) levels were detected by the enzyme-linked immunoassay method. After removing the adipose tissue, the obtained rat ovary tissue was used for subsequent experimental detection, using HE staining for morphology and follicular development analysis; qRT-PCR for the detection of 3ßHSD, CYP17α1, CYP19α1, and StAR gene expression in ovarian tissue; and western blotting analysis of CYP17α1, CYP19α1, StAR protein expression and insulin level to verify whether dulaglutide has a therapeutic effect on PCOS in rats. RESULTS: After treated with different concentrations of dulaglutide, we found that the body weight of rats in the treatment groups were reduced. Compared with the rats in PCOS group, the serum androgen level of rats in the treatment groups was significantly decreased, and the serum sex hormone binding protein content was significantly increased, and there was statistically significant difference between these groups and PCOS group. In terms of protein expression and gene regulation, the expression of 3ßHSD, CYP19α1 and StAR in the ovarian tissue of rats in treatment groups were decreased significantly after received the treatment of dulaglutide, and there was statistically significant difference between these groups and PCOS group. In addition, dulaglutide reduced the insulin content in the ovarian tissue of PCOS rats. CONCLUSION: Dulaglutide may reduce the hyperandrogenemia of PCOS rats by regulating the content of serum SHBG and the expression of 3ßHSD, CYP19α1, and StAR related genes and proteins, thereby inhibiting the excessive development of small follicles and the formation of cystic follicles in the ovaries of PCOS rats, thereby improving polycystic ovary in PCOS rats. In addition, dulaglutide may reduce the weight of PCOS rats, further reducing the level of high androgen in PCOS rats, and improving the morphology of their polycystic ovaries.


Asunto(s)
Péptidos Similares al Glucagón/análogos & derivados , Fragmentos Fc de Inmunoglobulinas/farmacología , Ovario/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Animales , Peso Corporal/efectos de los fármacos , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Péptidos Similares al Glucagón/farmacología , Resistencia a la Insulina , Ovario/fisiopatología , Ovulación/efectos de los fármacos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/fisiopatología , Ratas Sprague-Dawley , Globulina de Unión a Hormona Sexual/análisis , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo , Testosterona/sangre
4.
Life Sci ; 276: 119456, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33811895

RESUMEN

BACKGROUND: In recent years, female infertility from Polycystic Ovary Syndrome (PCOS) has gained scientific interest. PCOS alters the metabolic and endocrine functioning in females. The elevation in androgens can damage the androgen receptors present on the kidney giving rise to renal disorders like Focal Segmental Glomerulosclerosis (FSGS). Transforming Growth Factor Beta (TGF-ß) in the ovary is activated by activin for Follicle Stimulating Hormone (FSH) secretion and in the kidney by thrombospondin 1 (TSP1) for cell growth and apoptosis. Studies show that gamma-linolenic acid (GLA) effectively treats breast cancer, eczema, inflammatory conditions and PCOS. AIM: The study aimed to find out the possibility of FSGS development in PCOS and to understand the effect of GLA on FSGS via the TGF-ß pathway. METHOD: To carry out the study, the dehydroepiandrosterone (DHEA) induced PCOS model was used. Three groups namely vehicle control, DHEA, and DHEA+GLA, were used with six animals in each. TGF-ß1, TGF-ß2, and TSP1 genes were studied using real-time PCR. RESULTS: The study showed an increase in the level of renal fibrosis biomarker, TSP1, in the DHEA group, which was further decreased by an anti-inflammatory agent, GLA. The TGF-ß1 and TGF-ß2 genes associated with the TGF-ß pathway were seen to be increased in DHEA-induced PCOS rats which showed a possible relation between the two conditions. CONCLUSION: The study shows a possible development of renal fibrosis in the DHEA-induced PCOS model. The GLA might act as a ligand to regulate TGF-ß signaling in glomerulosclerosis in a DHEA-induced PCOS model.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Síndrome del Ovario Poliquístico/complicaciones , Factor de Crecimiento Transformador beta/metabolismo , Ácido gammalinolénico/farmacología , Adyuvantes Inmunológicos/toxicidad , Animales , Deshidroepiandrosterona/toxicidad , Femenino , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/patología , Ratas , Ratas Wistar , Transducción de Señal , Factor de Crecimiento Transformador beta/genética
5.
Reprod Sci ; 28(10): 2807-2815, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33876387

RESUMEN

Reactive oxygen species (ROS), involved in the pathogenesis of the polycystic ovary syndrome (PCOS), play a key role in the onset of apoptosis in follicles and granulosa cells (GCs). We aimed to investigate the antioxidant effects of AST and metformin separately and in combination on GCs using a PCOS mouse model. Forty-eight prepubertal female BALB C mice aged 25-30 days and weighing 12-14 g were studied. The PCOS model was created by subcutaneous injection of the dehydroepiandrosterone (DHEA) hormone in 8 mice of BALB C for 20 consecutive days. Apoptosis and the amount of ROS were evaluated in GCs of the ovaries via flow cytometry. The activity of AKT protein was measured by western blot, and the viability of GCs was investigated using spectrophotometry. Ovarian tissue sections were prepared, stained with H&E, and the morphology of the sections was examined. Statistical analysis was performed by SPSS v22.0 software using one-way ANOVA. We found that AST administration leads to a significant reduction in oxidative stress (P<0.01) and consequently a significant decrease in the rate of apoptosis (P<0.01). While the expression of AKT in the AST group revealed a significant increase (P<0.05), it decreased in the metformin group. However, it was still significantly higher than the control and PCOS groups. Ovulation was confirmed in both metformin and AST groups. Further studies are warranted to prove the efficacy of AST and to introduce it as a complementary therapeutic agent in PCOS.


Asunto(s)
Células de la Granulosa/efectos de los fármacos , Metformina/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Animales , Deshidroepiandrosterona/toxicidad , Femenino , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Metformina/farmacología , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/fisiología , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Xantófilas/farmacología , Xantófilas/uso terapéutico
6.
J Cell Mol Med ; 24(18): 11001-11011, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32808450

RESUMEN

Polycystic ovary syndrome (PCOS), the most common female endocrine disease that causes anovulatory infertility, still lacks promising strategy for the accurate diagnosis and effective therapeutics of PCOS attributed to its unclear aetiology. In this study, we determined the abnormal reduction in circPSMC3 expression by comparing the ovarian tissue samples of PCOS patients and normal individuals. The symptom relief caused by up-regulation of circPSMC3 in PCOS model mice suggested the potential for further study. In vitro functional experiments confirmed that circPSMC3 can inhibit cell proliferation and promote apoptosis by blocking the cell cycle in human-like granular tumour cell lines. Mechanism study revealed that circPSMC3 may play its role through sponging miR-296-3p to regulate PTEN expression. Collectively, we preliminarily characterized the role and possible insights of circPSMC3/miR-296-3p/PTEN axis in the proliferation and apoptosis of KGN cells. We hope that this work provides some original and valuable information for the research of circRNAs in PCOS, not only to better understand the pathogenesis but also to help provide new clues for seeking for the future therapeutic target of PCOS.


Asunto(s)
Terapia Genética , MicroARNs/genética , Fosfohidrolasa PTEN/biosíntesis , Síndrome del Ovario Poliquístico/terapia , ARN Circular/genética , Adulto , Animales , Apoptosis , Puntos de Control del Ciclo Celular , División Celular , Línea Celular Tumoral , Deshidroepiandrosterona/toxicidad , Femenino , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Tumor de Células de la Granulosa/patología , Células de la Granulosa/metabolismo , Humanos , Insulina/sangre , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Persona de Mediana Edad , Neoplasias Ováricas/patología , Ovario/metabolismo , Fosfohidrolasa PTEN/genética , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , ARN/metabolismo , ARN Circular/metabolismo , ARN Circular/uso terapéutico , Regulación hacia Arriba
7.
Artículo en Inglés | MEDLINE | ID: mdl-32160160

RESUMEN

Background It is estimated that about 5-10% of women suffer from polycystic ovarian syndrome (PCOS) which is a major cause of female reproductive dysfunction. This study examined the role of quercetin on dehydroepiandrosterone (DHEA)-induced PCO in Wistar rats. Methods Twenty-eight pre-pubertal female Wistar rats that are 21 days old weighing 16-21 g were sorted into four groups (n = 7). Group I served as control and was given distilled water only, Group II were injected with 6 mg/100 g BW of DHEA in 0.2 mL of corn oil subcutaneously, Group III received 100 mg/kg BW of quercetin orally and Group IV received 6 mg/100 g BW of DHEA in 0.2 mL of corn oil subcutaneously and 100 mg/kg BW of quercetin orally. Rats were sacrificed after 15 days by cervical dislocation method. Blood samples and ovaries were collected for hormonal, biochemical, and histopathological analysis and expressions of mRNA androgen receptor gene were determined using RT-qPCR. All data were analysed using one-way ANOVA. Results A significant decrease (p < 0.05) in the antioxidant and metabolic enzyme activity in the DHEA treated group was observed when compared with control. DHEA co-administration with quercetin showed a significant decrease in malondialdehyde and cytokines when compared with DHEA treated group. Also a significant increase in progesterone, metabolic and antioxidant enzyme activity was observed. The histopathology demonstrates a reduction in cystic and atretic cells, improved expression of BCl2, E-Cadherin and a decrease in Bax. Conclusions Quercetin alleviated DHEA-induced PCO. These effects could be attributed to its antioxidant property.


Asunto(s)
Células de la Granulosa/efectos de los fármacos , Ovario/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/genética , Quercetina/farmacología , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Adyuvantes Inmunológicos/toxicidad , Animales , Antioxidantes/farmacología , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores Androgénicos/genética
8.
J Cell Physiol ; 235(9): 6113-6126, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32048305

RESUMEN

Polycystic ovarian syndrome (PCOS) is a disorder characterized by oligomenorrhea, anovulation, and hyperandrogenism. Altered mitochondrial biogenesis can result in hyperandrogenism. The goal of this study was to examine the effect of vitamin D3 on mitochondrial biogenesis of the granulosa cells in the PCOS-induced mouse model. Vitamin D3 applies its effect via the mitogen-activated pathway kinase-extracellular signal-regulated kinases (MAPK-ERK1/2) pathway. The PCOS mouse model was induced by the injection of dehydroepiandrosterone (DHEA). Isolated granulosa cells were subsequently treated with vitamin D3, MAPK activator, and MAPK inhibitor. Gene expression levels were measured using real-time polymerase chain reaction. MAPK proteins were investigated by western blot analysis. We also determined reactive oxygen species (ROS) levels with 2', 7'-dichlorofluorescein diacetate. Mitochondrial membrane potential (mtMP) was also measured by TMJC1. Mitochondrial biogenesis (peroxisome proliferator-activated receptor gamma coactivator 1-α and nuclear respiratory factor), antioxidant (superoxide dismutase, glutathione peroxidase, and catalase), and antiapoptotic (B-cell lymphoma-2) genes were upregulated in the PCOS mice that treated with vitamin D3 compared with the PCOS mice without any treatment. Vitamin D3 and MAPK activator-treated groups also reduced ROS levels compared with the nontreated PCOS group. In summary, vitamin D3 and MAPK activator increased the levels of mitochondrial biogenesis, MAPK pathway, and mtMP markers, while concomitantly decreased ROS levels in granulosa cells of the PCOS-induced mice. This study suggests that vitamin D3 may improve mitochondrial biogenesis through stimulation of the MAPK pathway in cultured granulosa cells of DHEA-induced PCOS mice which yet to be investigated.


Asunto(s)
Colecalciferol/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Biogénesis de Organelos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Catalasa/genética , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Femenino , Glutatión Peroxidasa/genética , Células de la Granulosa/efectos de los fármacos , Humanos , Ratones , Factores Nucleares de Respiración/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/patología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/genética
9.
Curr Med Sci ; 38(5): 853-860, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30341520

RESUMEN

The polycystic ovary syndrome (PCOS) model was established in rats and correlation between the expression of macrophage migration inhibitory factor (MIF) and cytokinesis with the MAPK signalling pathway in the rat ovary was measured. The PCOS model in rats was established by dehydroepiandrosterone (DHEA). Thirty sexually immature female Sprague-Dawley rats were randomly and equally assigned to three groups: control group, PCOS group, and PCOS with high-fat diet (HFD) group. Serum hormones were assayed by radioimmunoassay (RIA). The ovaries were immunohistochemically stained with MIF, and the expression of MIF, p-JNK and p-p38 was detected by Western blotting in ovaries. The serum testosterone level, LH concentration, LH/FSH ratio, fasting insulin level and HOMA IR index in the PCOS group (6.077±0.478, 13.809±1.701, 1.820±0.404, 10.83±1.123 and 1.8692±0.1096) and PCOS with HFD group (6.075±0.439, 14.075±1.927, 1.779±0.277, 10.20±1.377 and 1.7736±0.6851) were significantly higher than those in the control group (4.949±0.337, 2.458±0.509, 1.239±0.038, 9.53±0.548 and 1.5329±0.7363), but there was no significant difference between the PCOS group and PCOS with HFD group. The expression levels of MIF, p-JNK, and p-p38 in the PCOS group (0.4048±0.013, 0.6233±0.093 and 0.7987±0.061) and PCOS with HFD group (0.1929±0.012, 0.3346±0.103 and 0.3468±0.031) were obviously higher than those in control group (0.2492±0.013, 0.3271±0.093 and 0.3393±0.061), but no significant difference was observed between PCOS group and PCOS with HFD group. It was suggested that MIF may participate in the pathogenesis of PCOS through the MAPK signalling pathway in PCOS rats induced by DHEA.


Asunto(s)
Resistencia a la Insulina/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Síndrome del Ovario Poliquístico/genética , Animales , Deshidroepiandrosterona/toxicidad , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ayuno , Femenino , Humanos , Hormona Luteinizante/sangre , Sistema de Señalización de MAP Quinasas/genética , Ovario/crecimiento & desarrollo , Ovario/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/fisiopatología , Ratas , Ratas Sprague-Dawley , Testosterona/sangre
10.
Ultrastruct Pathol ; 42(1): 55-64, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29192811

RESUMEN

AIM: The aim of this study was to investigate the effects of vitamin D treatment on ovary in experimentally designed polycystic ovary syndrome of female rats using light and electron microscopic techniques. METHODS: Twenty-four female pre-pubertal rats were divided into control, DHEA and DHEA+Vit.D groups. In DHEA group, the PCOS rat model was developed by 6mg/kg/day dehydroepiandrosterone administration as subcutaneously injections. In DHEA+Vit.D group, 6 mg/kg/day DHEA and 120ng/100g/week 1,25(OH)2D3 was performed simultaneously. Controls were injected with vehicle alone. At the end of the 28 days, blood samples were collected and the ovarian tissues were taken for histological examinations. RESULTS: FSH, LH levels, LH/FSH ratio, and testosterone levels showed a significant increase in DHEA group when compared with the control group. Moreover, these measurements were lower in the treatment group than the DHEA group. In DHEA group, increased number of atretic follicles and cystic follicles were seen with light microscopic analysis. Cystic follicles with attenuated granulosa cell layers and thickened theca cell layers and lipid accumulation in interstitial cells were observed by electron microscope. It is observed that atretic and cystic follicles were decreased as a result of vitamin D treatment. CONCLUSION: Our results indicate the curative role of vitamin D treatment on the androgen excess in PCOS rat model which causes abnormalities in ovarian morphology and functions. Vitamin D has positive effects on the hormonal and structural changes observed in PCOS, but it has been concluded that long-term use may be more beneficial.


Asunto(s)
Antioxidantes/farmacología , Ovario/ultraestructura , Síndrome del Ovario Poliquístico/patología , Vitamina D/farmacología , Animales , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Femenino , Microscopía Electrónica de Transmisión , Ovario/efectos de los fármacos , Ovario/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Ratas , Ratas Wistar
11.
Reproduction ; 152(6): 705-714, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27798284

RESUMEN

Polycystic ovary syndrome (PCOS) is a common reproductive disorder that has many characteristic features including hyperandrogenemia, insulin resistance and obesity, which may have significant implications for pregnancy outcomes and long-term health of women. Daughters born to PCOS mothers constitute a high-risk group for metabolic and reproductive derangements, but no report has described potential growth and metabolic risk factors for such female offspring. Hence, we used a mouse model of dehydroepiandrosterone (DHEA)-induced PCOS to study the mechanisms underlying the pathology of PCOS by investigating the growth, developmental characteristics, metabolic indexes and expression profiles of key genes of offspring born to the models. We found that the average litter size was significantly smaller in the DHEA group, and female offspring had sustained higher body weight, increased body fat and triglyceride content in serum and liver; they also exhibited decreased energy expenditure, oxygen consumption and impaired glucose tolerance. Genes related to glucolipid metabolism such as Pparγ, Acot1/2, Fgf21, Pdk4 and Inhbb were upregulated in the liver of the offspring in DHEA group compared with those in controls, whereas Cyp17a1 expression was significantly decreased. However, the expression of these genes was not detected in male offspring. Our results show that female offspring in DHEA group exhibit perturbed growth and glucolipid metabolism that were not observed in male offspring.


Asunto(s)
Deshidroepiandrosterona/toxicidad , Regulación de la Expresión Génica , Hígado/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Animales , Presión Sanguínea/efectos de los fármacos , Femenino , Prueba de Tolerancia a la Glucosa , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Síndrome del Ovario Poliquístico/inducido químicamente , Embarazo
12.
Biol Reprod ; 95(4): 79, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27557647

RESUMEN

Patients with polycystic ovary syndrome (PCOS) can suffer from psychological disorders, among which depression is the most commonly diagnosed. However, the pathogenesis is still unclear. The aims of the present study were to investigate the behaviors of dehydroepiandrosterone (DHEA)-induced PCOS mice, the effects of high-fat diet (HFD) on mouse behaviors, and the underlying mechanism. Prepubertal C57BL/6 mice (25 days of age) were divided into four groups and injected daily with the vehicle sesame oil or DHEA on the normal chow or a 60% HFD for 20 consecutive days. Depression-like behavior of the mice was examined using a forced swim test, tail suspension test, and open field test. Thereafter, the animals were killed and four brain regions were collected. The brain levels of monoamines and their metabolites, including norepinephrine, serotonin, 5-hydroxy-3-indolacetic acid, dopamine, and 3,4-hydroxyphenylacetic acid, were analyzed by HPLC. Our data show that DHEA-treated mice exhibited depression-like behavior according to the results from behavioral assessment. The brain contents of monoamines and/or their metabolites decreased in DHEA-treated mice compared with controls. HFD did not seem to markedly affect the behavioral responses, the brain monoamines, or their metabolites in the mice. These findings suggest that DHEA treatment induced depression-like behavior in PCOS mice, possibly through down-regulation of brain monoamines and/or their metabolites, which implies the contribution of hyperandrogenism to the psychological symptoms of women with PCOS.


Asunto(s)
Conducta Animal/efectos de los fármacos , Deshidroepiandrosterona/toxicidad , Depresión/etiología , Síndrome del Ovario Poliquístico/psicología , Animales , Conducta Animal/fisiología , Monoaminas Biogénicas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Depresión/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/etiología , Distribución Tisular
13.
Reproduction ; 151(4): 431-41, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26814210

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder with unclear etiology and unsatisfactory management. Effects of diets on the phenotype of PCOS were not fully understood. In the present study, we applied 45 and 60% high-fat diets (HFDs) on a rat model of PCOS induced by postnatal DHEA injection. We found that both DHEA and DHEA+HFDs rats exhibited reproductive abnormalities, including hyperandrogenism, irregular cycles and polycystic ovaries. The addition of HFDs, especially 60% HFDs, exaggerated morphological changes of ovaries and a number of metabolic changes, including increased body weight and body fat content, impaired glucose tolerance and increased serum insulin levels. Results from qPCR showed that DHEA-induced increased expression of hypothalamic androgen receptor and LH receptor were reversed by the addition of 60% HFDs. In contrast, the ovarian expression of LH receptor and insulin receptor mRNA was upregulated only with the addition of 60% HFDs. These findings indicated that DHEA and DHEA+HFDs might influence PCOS phenotypes through distinct mechanisms: DHEA affects the normal function of hypothalamus-pituitary-ovarian axis through LH, whereas the addition of HFDs exaggerated endocrine and metabolic dysfunction through ovarian responses to insulin-related mechanisms. We concluded that the addition of HFDs yielded distinct phenotypes of DHEA-induced PCOS and could be used for studies on both reproductive and metabolic features of the syndrome.


Asunto(s)
Deshidroepiandrosterona/toxicidad , Dieta Alta en Grasa/efectos adversos , Ciclo Estral/efectos de los fármacos , Resistencia a la Insulina , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Animales , Peso Corporal , Femenino , Prueba de Tolerancia a la Glucosa , Fenotipo , Síndrome del Ovario Poliquístico/etiología , Ratas , Ratas Sprague-Dawley
14.
Hum Exp Toxicol ; 35(5): 562-72, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26199281

RESUMEN

The aim of this study is to assess cardiotoxic effect of testosterone (TES) and dehydroepiandrosterone (DHEA) in Sprague Dawley rats. We compared the impact of subacute (14 days) and subchronic (90 days) administration of suprapharmacologic doses of TES and DHEA on body weight, locomotor activity, muscle strength, echocardiographic parameters, heart histopathology, and oxidative stress markers with the control group. Testosterone (10, 30, and 100 mg/100 g body weight) and DHEA (10 mg/100 g body weight) administration decreased the body weights and locomotor activity (p < 0.05), and the combination of both increased muscle strength (p < 0.05) in rats. In our histopathological evaluation, misshapen cell nuclei, disorganized myocardial fibers, and leukocytic infiltrates were observed in high-dose TES (100 mg/100 g)-treated rats, especially on day 14. On day 90, mild changes such as misshapen cell nuclei, disorganized myocardial fibers, and leukocytic infiltrates were observed in TES and DHEA-treated groups. According to our echocardiographic study on day 14 and day 90, TES, especially at high doses, induced increase in left ventricular posterior wall diameter and ejection fraction (p < 0.05). In this study, blood oxidative stress marker malondialdehyde was increased slightly but not significantly in TES and DHEA groups. On the other hand, antioxidant enzymes such as SOD and glutathione peroxidase (GSH-Px) levels were slightly but not significantly increased in TES and DHEA groups. These data demonstrate that the potential risk to cardiac health due to exogenous androgen use may be related to oxidative stress in rats.


Asunto(s)
Andrógenos/toxicidad , Deshidroepiandrosterona/toxicidad , Corazón/efectos de los fármacos , Miocardio , Estrés Oxidativo/efectos de los fármacos , Testosterona/toxicidad , Andrógenos/administración & dosificación , Animales , Peso Corporal/efectos de los fármacos , Cardiotoxicidad , Deshidroepiandrosterona/administración & dosificación , Relación Dosis-Respuesta a Droga , Ecocardiografía , Masculino , Actividad Motora/efectos de los fármacos , Fuerza Muscular/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Ratas Wistar , Testosterona/administración & dosificación
15.
J Ovarian Res ; 8: 64, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26437930

RESUMEN

BACKGROUND: Polycystic Ovary Syndrome (PCOS) is a widespread reproductive disorder characterized by a disruption of follicular growth and anovulatory infertility. In women with PCOS, follicular growth and ovulation can be induced by subcutaneous injections of low doses of follicle stimulating hormone (FSH). The aim of this study was to determine the effect of oral administration of recombinant human FSH (rhFSH) on follicle development in a PCOS murine model. Moreover, since it is unlikely that intact rhFSH is present into the circulation after oral administration, the biological activity of a peptide fragment, derived from the predicted enzymatic cleavage sites with the FSH molecule, was investigated in vitro on cumulus-enclosed oocytes (COCs). METHODS: Female peripubertal mice were injected with dehydroepiandrosterone (DHEA) diluted in sesame oil for 20 consecutive days and orally treated with a saline solution of rhFSH. A control group received only sesame oil and saline solution. At the end of treatments, blood was analyzed for hormone concentrations and ovaries were processed for morphological analysis. The presumptive bioactive peptide was added during in vitro maturation of bovine COCs and the effects on cumulus expansion and on maturation rate were evaluated. RESULTS: DHEA treatment increased serum levels of testosterone, estradiol and progesterone as well as the percentage of cystic follicles. Orally administered rhFSH restored estradiol level and reduced the percentage of cystic follicles. Despite these results indicating a reduction of the severity of PCOS in the mouse model, the presumptive bioactive peptide did not mimic the effect of rhFSH and failed to induce bovine cumulus expansion and oocyte maturation in vitro. CONCLUSIONS: Although further studies are needed, the present data supports the concept that orally administrated FSH could attenuate some of the characteristic of PCOS in the mouse model.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Adyuvantes Inmunológicos/toxicidad , Administración Oral , Análisis de Varianza , Animales , Aromatasa/metabolismo , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Estradiol/metabolismo , Femenino , Hormona Folículo Estimulante/administración & dosificación , Hormonas Esteroides Gonadales/metabolismo , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/tratamiento farmacológico , Inyecciones Intradérmicas , Ratones Endogámicos BALB C , Oocitos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Ovario/efectos de los fármacos , Proteínas Recombinantes , Aumento de Peso
16.
Eur J Med Chem ; 68: 301-11, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23994323

RESUMEN

The aim of this study was to determine the cytotoxic effect of human cancer cells on three series of novel dehydroepiandrosterone derivatives containing triazole or pyrazole rings at C-17 and an ester moiety at C-3 of the androstane skeleton. The panel cancer cells used in this study were the following: PC-3, MCF-7 and SKLU-1. The results from this study indicated that the steroidal derivatives 4a-4e and 4f-4k showed the highest cytotoxic potency. This difference in this activity could be attributed to the ability of the triazole (three nitrogen atoms) to form stronger hydrogen bonds with the active site of the cell as compared to the pyrazole group having two nitrogen atoms. Compounds 4f-4k having an aromatic ester at C-3 showed an enhanced cytotoxic activity as compared to their aliphatic counterparts 4a-4e. Apparently the electronegative phenyl ring increased the polarity of the molecule, thus increasing the dipole-dipole association of the steroidal molecule with the reactive site of the cell.


Asunto(s)
Deshidroepiandrosterona/toxicidad , Neoplasias/tratamiento farmacológico , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Deshidroepiandrosterona/síntesis química , Deshidroepiandrosterona/química , Humanos , Relación Estructura-Actividad
17.
Artículo en Inglés | MEDLINE | ID: mdl-21704190

RESUMEN

Rainbow trout (Oncorhynchus mykiss) are an outstanding model of liver cancer induction by environmental chemicals and development of strategies for chemoprevention. Trout have critical and unique advantages allowing for cancer studies with 40,000 animals to determine dose-response at levels orders of magnitude lower than possible in rodents. Examples of two promoters in this model, the dietary supplement dehydroepiandrosterone (DHEA) and industrial chemical perfluorooctanoic acid (PFOA), are presented. In addition, indole-3-carbinol (I3C) and chlorophyllin (CHL) inhibit initiation following exposure to potent human chemical carcinogens (e.g., aflatoxin B(1) (AFB(1))). Two "ED(001)" cancer studies have been conducted, utilizing approximately 40,000 trout, by dietary exposure to AFB(1) and dibenzo[d,e,f,p]chrysene (DBC). These studies represent the two largest cancer studies ever performed and expand the dose-response dataset generated by the 25,000 mouse "ED(01)" study over an order of magnitude. With DBC, the liver tumor response fell well below the LED(10) line, often used for risk assessment, even though the biomarker (liver DBC-DNA adducts) remained linear. Conversely, the response with AFB(1) remained relatively linear throughout the entire dose range. These contributions to elucidation of mechanisms of liver cancer, induced by environmental chemicals and the remarkable datasets generated with ED(001) studies, make important contributions to carcinogenesis and chemoprevention.


Asunto(s)
Quimioprevención , Exposición a Riesgos Ambientales/análisis , Neoplasias Hepáticas Experimentales/inducido químicamente , Aflatoxina B1/toxicidad , Animales , Benzopirenos/toxicidad , Caprilatos/toxicidad , Clorofilidas/farmacología , Clorofilidas/uso terapéutico , Deshidroepiandrosterona/toxicidad , Relación Dosis-Respuesta a Droga , Fluorocarburos/toxicidad , Indoles/farmacología , Indoles/uso terapéutico , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/prevención & control , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/fisiología , Regiones Promotoras Genéticas , Medición de Riesgo
18.
J Cereb Blood Flow Metab ; 29(2): 287-96, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18854841

RESUMEN

Dehydroepiandrosterone (DHEA) has been implicated not only to prevent N-methyl-D-aspartate (NMDA)-induced neurotoxicity but also to enhance Ca(2+) influx through NMDA receptor (NMDAr). However, these DHEA effects, which would produce inconsistent outcomes about neuronal damages, are not well studied in ischemia-induced cerebral damages. Herein, we report that a single administration of DHEA (20 mg/kg) during 3 to 48 h after transient global cerebral ischemia in rats exerted neuroprotective effects such as reduction of ischemia-induced neuronal death in the hippocampal CA1 and improvement of ischemia-induced deficits in spatial learning. By contrast, at 1 h before or after ischemia, the administration of DHEA exacerbated the ischemia-induced neuronal death and learning impairment. This DHEA neurotoxicity appeared to be caused by DHEA itself, but not through its metabolite testosterone, and was inhibited by a pretreatment with the NMDAr blocker MK801 or the sigma-1 (sigma(1)) receptor antagonist NE100. However, the DHEA neuroprotection was blocked by NE100. These results show that DHEA not only provides robust ischemic neuroprotection with a long therapeutic opportunity but also exerts neurotoxicity when administered during ischemia and early reperfusion, which points to the importance of administration timing of DHEA in the clinical treatment of brain damages by the transient brain ischemia including stroke.


Asunto(s)
Deshidroepiandrosterona/toxicidad , Deshidroepiandrosterona/uso terapéutico , Ataque Isquémico Transitorio/tratamiento farmacológico , Fármacos Neuroprotectores/toxicidad , Fármacos Neuroprotectores/uso terapéutico , Animales , Muerte Celular , Ataque Isquémico Transitorio/patología , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo
19.
Int Immunopharmacol ; 8(13-14): 1827-34, 2008 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-18817896

RESUMEN

The aim of the present work was to study some of the adverse effects produced by hyperandrogenism on the uterine function. Daily injection of dehydroepiandrosterone (DHEA: 6 mg/ 100 g body weight, sc) for 20 consecutive days induced polycystic ovaries in BALB/c mice. In this model, we found that DHEA produced alterations on uterine histology closely related to the development of tumour structures. In addition, hyperandrogenism induced a pro-inflammatory and a pro-oxidant condition represented by increased levels of prostaglandin F2 alpha production and uterine nitric oxide synthase (NOS) activity and by a decrease in both superoxide dismutase (SOD) and catalase (CAT) activities together with a decrease in the levels of the antioxidant metabolite glutathione (GSH). DHEA also induced an increase in CD4+ together with a decrease in the CD8+ T lymphocytes that infiltrate the uterine tissue. We conclude that this intricate network of regulators could be responsible for the low rate of implantation observed in women with polycystic ovary syndrome.


Asunto(s)
Adyuvantes Inmunológicos/toxicidad , Deshidroepiandrosterona/toxicidad , Hiperandrogenismo/fisiopatología , Síndrome del Ovario Poliquístico/inducido químicamente , Útero/fisiopatología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Catalasa/antagonistas & inhibidores , Catalasa/metabolismo , Ciclooxigenasa 1/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Dinoprost/biosíntesis , Femenino , Glutatión/antagonistas & inhibidores , Glutatión/metabolismo , Hiperandrogenismo/patología , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa/efectos de los fármacos , Óxido Nítrico Sintasa/metabolismo , Ovario/patología , Estrés Oxidativo/efectos de los fármacos , Síndrome del Ovario Poliquístico/inmunología , Síndrome del Ovario Poliquístico/patología , Prostaglandinas E/biosíntesis , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo , Útero/inmunología , Útero/patología
20.
Environ Health Perspect ; 116(8): 1047-55, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18709148

RESUMEN

BACKGROUND: Perfluorooctanoic acid (PFOA) is a potent hepatocarcinogen and peroxisome proliferator (PP) in rodents. Humans are not susceptible to peroxisome proliferation and are considered refractory to carcinogenesis by PPs. Previous studies with rainbow trout indicate they are also insensitive to peroxisome proliferation by the PP dehydroepiandrosterone (DHEA), but are still susceptible to enhanced hepatocarcinogenesis after chronic exposure. OBJECTIVES: In this study, we used trout as a unique in vivo tumor model to study the potential for PFOA carcinogenesis in the absence of peroxisome proliferation compared with the structurally diverse PPs clofibrate (CLOF) and DHEA. Mechanisms of carcinogenesis were identified from hepatic gene expression profiles phenotypically anchored to tumor outcome. METHODS: We fed aflatoxin B(1) or sham-initiated animals 200-1,800 ppm PFOA in the diet for 30 weeks for tumor analysis. We subsequently examined gene expression by cDNA array in animals fed PFOA, DHEA, CLOF, or 5 ppm 17beta-estradiol (E(2), a known tumor promoter) in the diet for 14 days. RESULTS: PFOA (1,800 ppm or 50 mg/kg/day) and DHEA treatments resulted in enhanced liver tumor incidence and multiplicity (p < 0.0001), whereas CLOF showed no effect. Carcinogenesis was independent of peroxisome proliferation, measured by lack of peroxisomal beta-oxidation and catalase activity. Alternately, both tumor promoters, PFOA and DHEA, resulted in estrogenic gene signatures with strong correlation to E(2) by Pearson correlation (R = 0.81 and 0.78, respectively), whereas CLOF regulated no genes in common with E(2). CONCLUSIONS: These data suggest that the tumor-promoting activities of PFOA in trout are due to novel mechanisms involving estrogenic signaling and are independent of peroxisome proliferation.


Asunto(s)
Caprilatos/toxicidad , Fluorocarburos/toxicidad , Genómica , Neoplasias Hepáticas/inducido químicamente , Oncorhynchus mykiss/genética , Peroxisomas/efectos de los fármacos , Aflatoxina B1/toxicidad , Animales , Pruebas de Carcinogenicidad , Clofibrato/toxicidad , Análisis por Conglomerados , Deshidroepiandrosterona/toxicidad , Modelos Animales de Enfermedad , Estradiol/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncorhynchus mykiss/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA