Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Reprod Sci ; 27(10): 1909-1919, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32519158

RESUMEN

Placental function is of utmost importance to ensure proper fetal development in utero. Among the placenta's many roles includes the passage of sufficient macronutrients, such as glucose, amino acids, and fatty acids, to the fetus. Macronutrients are carried from maternal circulation to the fetus across transporters within the placenta. The objective of this study was to examine the impact of (i) an acute bout of exercise and (ii) chronic exercise participation on placenta nutrient transporter expression and localization. To investigate the effect of acute exercise, pre- and post-exercise serum was collected from pregnant (n = 5) and non-pregnant (n = 5) women who underwent a moderate-intensity exercise session and used to treat BeWo cells. To assess chronic physical activity, we analyzed term placenta from women categorized as active (n = 10) versus non-active (n = 10). Protein expression and localization for the transporters GLUT1, SNAT1, and FATP4 were examined for both groups. GLUT1 expression in BeWo cells treated with serum from pregnant women was higher compared with that from non-pregnant, independent of exercise. FATP4 protein expression was elevated in the term placenta of active women. Immunohistochemistry analysis of term placenta illustrated increased staining of FATP4 in placental tissue from active women and differential staining pattern of GLUT1 depending on physical activity status. Chronic exercise during pregnancy increases the expression of placental FATP4 in vivo, suggesting greater metabolism and usage of fatty acids. Additionally, serum from pregnant women could contain factors that increase GLUT1 protein expression in vitro. BeWo cells treated with pre- and post-exercise serum from pregnant women resulted in greater GLUT1 expression compared with those treated with pre- and post-exercise serum from non-pregnant women. Physical activity appears to differentially impact key placental transporters involved in the transfer and availability of nutrients from mother to fetus. Future research ought to examine the mechanisms involved in regulating these changes and their impact on fetal growth and health.


Asunto(s)
Ejercicio Físico/fisiología , Proteínas de Transporte de Ácidos Grasos/metabolismo , Placenta/metabolismo , Adulto , Sistema de Transporte de Aminoácidos A/metabolismo , Línea Celular , Femenino , Desarrollo Fetal , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Embarazo , Trofoblastos/metabolismo
2.
Biol Reprod ; 101(5): 961-974, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31347667

RESUMEN

The ovarian surface epithelium (OSE) is a monolayer of cells surrounding the ovary that is ruptured during ovulation. After ovulation, the wound is repaired, however, this process is poorly understood. In epithelial tissues, wound repair is mediated by an epithelial-to-mesenchymal transition (EMT). Transforming Growth Factor Beta-1 (TGFß1) is a cytokine commonly known to induce an EMT and is present throughout the ovarian microenvironment. We, therefore, hypothesized that TGFß1 induces an EMT in OSE cells and activates signaling pathways important for wound repair. Treating primary cultures of mouse OSE cells with TGFß1 induced an EMT mediated by TGFßRI signaling. The transcription factor Snail was the only EMT-associated transcription factor increased by TGFß1 and, when overexpressed, was shown to increase OSE cell migration. A polymerase chain reaction array of TGFß signaling targets determined Cyclooxygenase-2 (Cox2) to be most highly induced by TGFß1. Constitutive Cox2 expression modestly increased migration and robustly enhanced cell survival, under stress conditions similar to those observed during wound repair. The increase in Snail and Cox2 expression with TGFß1 was reproduced in human OSE cultures, suggesting these responses are conserved between mouse and human. Finally, the induction of Cox2 expression in OSE cells during ovulatory wound repair was shown in vivo, suggesting TGFß1 increases Cox2 to promote wound repair by enhancing cell survival. These data support that TGFß1 promotes ovulatory wound repair by induction of an EMT and activation of a COX2-mediated pro-survival pathway. Understanding ovulatory wound repair may give insight into why ovulation is the primary non-hereditary risk factor for ovarian cancer.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Ovario/fisiología , Cicatrización de Heridas , Animales , Supervivencia Celular , Ciclooxigenasa 2/genética , Dinoprostona/genética , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica , Ratones , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
3.
PLoS Genet ; 14(11): e1007788, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30418965

RESUMEN

Estrogen therapy increases the risk of ovarian cancer and exogenous estradiol accelerates the onset of ovarian cancer in mouse models. Both in vivo and in vitro, ovarian surface epithelial (OSE) cells exposed to estradiol develop a subpopulation that loses cell polarity, contact inhibition, and forms multi-layered foci of dysplastic cells with increased susceptibility to transformation. Here, we use single-cell RNA-sequencing to characterize this dysplastic subpopulation and identify the transcriptional dynamics involved in its emergence. Estradiol-treated cells were characterized by up-regulation of genes associated with proliferation, metabolism, and survival pathways. Pseudotemporal ordering revealed that OSE cells occupy a largely linear phenotypic spectrum that, in estradiol-treated cells, diverges towards cell state consistent with the dysplastic population. This divergence is characterized by the activation of various cancer-associated pathways including an increase in Greb1 which was validated in fallopian tube epithelium and human ovarian cancers. Taken together, this work reveals possible mechanisms by which estradiol increases epithelial cell susceptibility to tumour initiation.


Asunto(s)
Estradiol/efectos adversos , Ovario/efectos de los fármacos , Ovario/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Inhibición de Contacto/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Femenino , Humanos , Proteínas de la Membrana , Ratones , Ovario/patología , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual
4.
Sci Rep ; 7(1): 16702, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29196616

RESUMEN

Estrogen replacement therapy increases the risk of human ovarian cancer and exogenous estradiol accelerates the onset of ovarian cancer in mouse models. This study uses primary cultures of mouse ovarian surface epithelium (OSE) to demonstrate that one possible mechanism by which estrogen accelerates the initiation of ovarian cancer is by up-regulation of microRNA-378 via the ESR1 pathway to result in the down-regulation of a tumour suppressor called Disabled-2 (Dab2). Estrogen suppression of Dab2 was reproducible in vivo and across many cell types including mouse oviductal epithelium and primary cultures of human ovarian cancer cells. Suppression of Dab2 resulted in increased proliferation, loss of contact inhibition, morphological dysplasia, and resistance to oncogene-induced senescence - all factors that can sensitize OSE to transformation. Given that DAB2 is highly expressed in healthy human OSE and is absent in the majority of ovarian tumours, this study has taken the first steps to provide a mechanistic explanation for how estrogen therapy may play a role in the initiation of ovarian cancer.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Carcinoma Epitelial de Ovario/metabolismo , Transformación Celular Neoplásica , Células Epiteliales/metabolismo , Estradiol/efectos adversos , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Proteínas Reguladoras de la Apoptosis , Carcinoma Epitelial de Ovario/inducido químicamente , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Epitelio/metabolismo , Epitelio/patología , Estradiol/farmacología , Femenino , Humanos , Ratones , Ratones Noqueados , Neoplasias Ováricas/inducido químicamente , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ovario/patología , Proteínas Supresoras de Tumor/genética
5.
J Biophotonics ; 10(10): 1327-1334, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28009133

RESUMEN

Despite the many advances intended to enhance the response to treatment, the survival rate of patients with ovarian cancer has only marginally improved in the past few decades. One major cause for this, is the lack of diagnostics for platinum-resistant disease. The goal of this study was to determine whether Raman micro-spectroscopy in conjunction with multivariate statistical analysis could discriminate between chemically fixed cisplatin-resistant (A2780cp) and cisplatin-sensitive (A2780s) human ovarian carcinoma cells. Raman spectra collected from individual cells were pre-processed and subsequently analyzed with Principal Component Analysis - Linear Discriminant Analysis (PCA-LDA). Statistically significant differences (P <  0.0001) were observed between the Raman spectra of A2780s and A2780cp cells. A diagnostic accuracy of 82% was obtained using the PCA-LDA classifier model for the discrimination between the A2780s and A2780cp cells. The loading plot analysis suggests that relative increases in proteins and glutathione in the cisplatin-resistant cells compared to the cisplatin-sensitive cells are most likely the major source of discrimination between the two types of cells. These results support the potential application of Raman spectroscopy in the identification of chemo-resistant tumors prior to treatment.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias Ováricas/patología , Espectrometría Raman , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Curva ROC
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...