Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Biol Reprod ; 107(2): 574-589, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35377412

RESUMEN

Paternal obesity predisposes offspring to metabolic dysfunction, but the underlying mechanisms remain unclear. We investigated whether this metabolic dysfunction is associated with changes in placental vascular development and is fueled by endoplasmic reticulum (ER) stress-mediated changes in fetal hepatic development. We also determined whether paternal obesity indirectly affects the in utero environment by disrupting maternal metabolic adaptations to pregnancy. Male mice fed a standard chow or high fat diet (60%kcal fat) for 8-10 weeks were time-mated with female mice to generate pregnancies and offspring. Glucose tolerance was evaluated in dams at mid-gestation (embryonic day (E) 14.5) and late gestation (E18.5). Hypoxia, angiogenesis, endocrine function, macronutrient transport, and ER stress markers were evaluated in E14.5 and E18.5 placentae and/or fetal livers. Maternal glucose tolerance was assessed at E14.5 and E18.5. Metabolic parameters were assessed in offspring at ~60 days of age. Paternal obesity did not alter maternal glucose tolerance but induced placental hypoxia and altered placental angiogenic markers, with the most pronounced effects in female placentae. Paternal obesity increased ER stress-related protein levels (ATF6 and PERK) in the fetal liver and altered hepatic expression of gluconeogenic factors at E18.5. Offspring of obese fathers were glucose intolerant and had impaired whole-body energy metabolism, with more pronounced effects in female offspring. Metabolic deficits in offspring due to paternal obesity may be mediated by sex-specific changes in placental vessel structure and integrity that contribute to placental hypoxia and may lead to poor fetal oxygenation and impairments in fetal metabolic signaling pathways in the liver.


Asunto(s)
Obesidad , Placenta , Animales , Dieta Alta en Grasa/efectos adversos , Padre , Femenino , Glucosa/metabolismo , Humanos , Hipoxia/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Placenta/metabolismo , Placentación , Embarazo
2.
J Physiol ; 597(12): 3029-3051, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31081119

RESUMEN

KEY POINTS: Maternal obesity has been associated with shifts in intestinal microbiota, which may contribute to impaired barrier function Impaired barrier function may expose the placenta and fetus to pro-inflammatory mediators We investigated the impacts of diet-induced obesity in mice on maternal and fetal intestinal structure and placental vascularization Diet-induced obesity decreased maternal intestinal short chain fatty acids and their receptors, impaired gut barrier integrity and was associated with fetal intestinal inflammation. Placenta from obese mothers showed blood vessel immaturity, hypoxia, increased transcript levels of inflammation, autophagy and altered levels of endoplasmic reticulum stress markers. These data suggest that maternal intestinal changes probably contribute to adverse placental adaptations and also impart an increased risk of obesity in the offspring via alterations in fetal gut development. ABSTRACT: Shifts in maternal intestinal microbiota have been implicated in metabolic adaptations to pregnancy. In the present study, we generated cohorts of female C57BL/6J mice fed a control (17% kcal fat, n = 10-14) or a high-fat diet (HFD 60% kcal from fat, n = 10-14; ad libitum) aiming to investigate the impact on the maternal gut microbiota, intestinal inflammation and gut barrier integrity, placental inflammation and fetal intestinal development at embryonic day 18.5. HFD was associated with decreased relative abundances of short-chain fatty acid (SCFA) producing genera during pregnancy. These diet-induced shifts paralleled decreased maternal intestinal mRNA levels of SCFA receptor Gpr41, modestly decreased cecal butyrate, and altered mRNA levels of inflammatory cytokines and immune cell markers in the maternal intestine. Maternal HFD resulted in impaired gut barrier integrity, with corresponding increases in circulating maternal levels of lipopolysaccharide (LPS) and tumour necrosis factor. Placentas from HFD dams demonstrated blood vessel immaturity and hypoxia; decreased free carnitine, acylcarnitine derivatives and trimethylamine-N-oxide; and altered mRNA levels of inflammation, autophagy, and ER stress markers. HFD exposed fetuses had increased activation of nuclear factor-kappa B and inhibition of the unfolded protein response in the developing intestine. Taken together, these data suggest that HFD intake prior to and during pregnancy shifts the composition of the maternal gut microbiota and impairs gut barrier integrity, resulting in increased maternal circulating LPS, which may ultimate contribute to changes in placental vascularization and fetal gut development.


Asunto(s)
Dieta Alta en Grasa , Microbioma Gastrointestinal , Hipoxia , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Obesidad , Placenta/irrigación sanguínea , Animales , Femenino , Desarrollo Fetal , Feto , Hipoxia/metabolismo , Hipoxia/microbiología , Hipoxia/fisiopatología , Mucosa Intestinal/microbiología , Lipopolisacáridos/metabolismo , Masculino , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/microbiología , Obesidad/fisiopatología , Placenta/metabolismo , Embarazo
3.
Biol Reprod ; 98(5): 664-682, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351580

RESUMEN

Reproductive abnormalities are included as health complications in offspring exposed to poor prenatal nutrition. We have previously shown in a rodent model that offspring born to nutrient restriction during pregnancy are born small, enter puberty early, and display characteristics of early ovarian aging as adults. The present study investigated whether key proteins involved in follicle recruitment and growth mediate ovarian follicle loss. Pregnant rats were randomized to a standard diet throughout pregnancy and lactation (CON), or a calorie-restricted (50% of control) diet (UN) during pregnancy. Offspring reproductive phenotype was investigated at postnatal days 4, 27, and 65. Maternal UN resulted in young adult (P65) irregular estrous cyclicity due to persistent estrus, a significant loss of antral follicles, corpora lutea, and an increase in atretic follicles. This decrease in growing follicles in UN offspring appears to be due to increased apoptosis as seen by immunopositive staining of pro-apoptotic factor CASP3 (caspase 3) in ovaries of young adult offspring. UN prepubertal offspring had reduced expression levels of Fshr in antral follicles, which may contribute to a decrease in PI3K/AKT activation evident as a decrease in pAKT immunolocalization in prepubertal antral follicles. Moreover, neonatal ovaries of UN offspring show decreased levels of immunopositive staining for AMHR2 (anti-mullerian hormone receptor 2). Collectively, these data demonstrate that maternal UN during pregnancy impacts ovarian function in offspring as early as P65 and provides a model for understanding the mechanisms driving early life UN-induced follicle loss and reproductive dysfunction.


Asunto(s)
Restricción Calórica , Ciclo Estral/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Folículo Ovárico/metabolismo , Ovario/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Reproducción/fisiología , Animales , Apoptosis/fisiología , Caspasa 3/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptores de HFE/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología
4.
Biol Reprod ; 92(4): 110, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25810471

RESUMEN

Maternal nutritional restriction has been shown to induce impairments in a number of organ systems including the ovary. We have previously shown that maternal undernutrition induces fetal growth restriction and low birth weight, and results in an offspring ovarian phenotype characteristic of premature ovarian aging with reduced ovarian reserve. In the present study, we set out to investigate the underlying mechanisms that lead offspring of undernourished mothers to premature ovarian aging. Pregnant dams were randomized to 1) a standard diet throughout pregnancy and lactation (control), 2) a calorie-restricted (50% of control) diet during pregnancy, 3) a calorie-restricted (50% of control) diet during pregnancy and lactation, or 4) a calorie-restricted (50% of control) diet during lactation alone. The present study shows that early life undernutrition-induced reduction of adult ovarian follicles may be mediated by increased ovarian endoplasmic reticulum stress in a manner that increased follicular apoptosis but not autophagy. These changes were associated with a loss of ovarian vessel density and are consistent with an accelerated ovarian aging phenotype. Whether these changes are mediated specifically by a reduction in the local antioxidant environment is unclear, although our data suggest the possibility that ovarian melatonin may play a part in early life nutritional undernutrition and impaired offspring folliculogenesis.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Desnutrición/fisiopatología , Neovascularización Fisiológica , Ovario/irrigación sanguínea , Animales , Antioxidantes/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Beclina-1 , Restricción Calórica , Femenino , Fenómenos Fisiologicos Nutricionales Maternos , Folículo Ovárico/metabolismo , Folículo Ovárico/fisiología , Ovario/crecimiento & desarrollo , Embarazo , Ratas , Ratas Wistar , Flujo Sanguíneo Regional , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
PLoS One ; 19(7): e0307178, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39028700

RESUMEN

Women diagnosed with ovarian cancer frequently have a poor prognosis as their cancer is often diagnosed at more advanced stages when the cancer has metastasized. At this point surgery cannot remove all the tumor cells and while ovarian cancer cells often initially respond to chemotherapeutic agents like carboplatin and paclitaxel, resistance to these agents frequently occurs. Thus, novel therapies are required for the treatment of advanced stage ovarian cancer. One therapeutic option being explored is the regulation of non-coding RNAs such as microRNAs. An advantage of microRNAs is that they can regulate tens, hundreds and sometimes thousands of mRNAs in cells and thus may be more effective than chemotherapeutic agents or targeted therapies. To investigate the therapeutic potential of miR-200s in ovarian cancer, lentiviral vectors were used to overexpress both miR-200 clusters in two murine ovarian cancer cell lines, ID8 and 28-2. Overexpression of miR-200s reduced the expression of several mesenchymal genes and proteins, significantly inhibited proliferation as assessed by BrdU flow cytometry and significantly reduced invasion through Matrigel coated transwell inserts in both cell lines. Overexpression of miR-200s also increased basal apoptosis approximately 3-fold in both cell lines as determined by annexin V flow cytometry. Pathway analysis of RNA sequencing of control and miR-200 overexpressing ovarian cancer cells revealed that genes regulated by miR-200s were involved in processes like epithelial mesenchymal transition (EMT) and cell migration. Therefore, miR-200s can inhibit proliferation and increase apoptosis while suppressing tumor cell invasion and thus simultaneously target three key cancer pathways.


Asunto(s)
Apoptosis , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , MicroARNs , Invasividad Neoplásica , Neoplasias Ováricas , MicroARNs/genética , MicroARNs/metabolismo , Femenino , Animales , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Apoptosis/genética , Proliferación Celular/genética , Ratones , Línea Celular Tumoral , Humanos , Movimiento Celular/genética
6.
Sci Rep ; 9(1): 17621, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31772245

RESUMEN

We investigated whether diet-induced changes in the maternal intestinal microbiota were associated with changes in bacterial metabolites and their receptors, intestinal inflammation, and placental inflammation at mid-gestation (E14.5) in female mice fed a control (17% kcal fat, n = 7) or a high-fat diet (HFD 60% kcal fat, n = 9; ad libitum) before and during pregnancy. Maternal diet-induced obesity (mDIO) resulted in a reduction in maternal fecal short-chain fatty acid producing Lachnospiraceae, lower cecal butyrate, intestinal antimicrobial peptide levels, and intestinal SCFA receptor Ffar3, Ffar2 and Hcar2 transcript levels. mDIO increased maternal intestinal pro-inflammatory NFκB activity, colonic CD3+ T cell number, and placental inflammation. Maternal obesity was associated with placental hypoxia, increased angiogenesis, and increased transcript levels of glucose and amino acid transporters. Maternal and fetal markers of gluconeogenic capacity were decreased in pregnancies complicated by obesity. We show that mDIO impairs bacterial metabolite signaling pathways in the mother at mid-gestation, which was associated with significant structural changes in placental blood vessels, likely as a result of placental hypoxia. It is likely that maternal intestinal changes contribute to adverse maternal and placental adaptations that, via alterations in fetal hepatic glucose handling, may impart increased risk of metabolic dysfunction in offspring.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Feto/metabolismo , Glucosa/metabolismo , Intestinos/patología , Obesidad/metabolismo , Placenta/metabolismo , Complicaciones del Embarazo/metabolismo , Animales , Butiratos/metabolismo , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Hipoxia de la Célula , Citocinas/metabolismo , Ácidos Grasos Volátiles/análisis , Heces/química , Femenino , Microbioma Gastrointestinal , Regulación de la Expresión Génica , Edad Gestacional , Gluconeogénesis , Inflamación , Intestinos/microbiología , Macrófagos/fisiología , Ratones , Obesidad/etiología , Placenta/irrigación sanguínea , Placenta/patología , Embarazo , Receptores Toll-Like/metabolismo , beta-Defensinas/metabolismo
7.
Mol Ther Methods Clin Dev ; 9: 181-191, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29556508

RESUMEN

Newcastle disease virus (NDV) is a single-stranded, negative-sense RNA virus in the Paramyxoviridae family. Although primarily an avian pathogen, NDV is a potent oncolytic virus that has been shown to be safe and effective in a variety of preclinical cancer models and human clinical trials. To produce virus for oncolytic trials, NDV is commonly amplified in embryonated chicken eggs and purified from the allantoic fluid. Conventional methods for purifying virus from allantoic fluid often result in relatively low-titer preparations containing high levels of impurities, including immunogenic chicken host cell proteins from allantoic fluid. However, large quantities of virus need to be delivered intravenously to administer oncolytic NDV systemically to mice. This route of administration requires virus preparations that are both highly concentrated (to enable delivery of small volumes) and highly pure (to limit toxic effects from contaminants). Given the accumulation of promising preclinical and clinical data demonstrating the efficacy of NDV as an oncolytic agent, strategies for increasing the titer and purity of NDV preparations are sorely needed to allow for effective intravenous administration in mice. Here, we describe an optimized protocol for the rescue, production, and purification of high-titer in vivo-grade NDV for preclinical studies in mouse models.

8.
Reprod Sci ; 21(5): 648-57, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24155067

RESUMEN

Maternal obesity is associated with an increased risk of a number of pregnancy complications, including fetal demise, which may be linked to impaired placental development as a result of altered trophoblast invasion and vessel remodeling. Therefore, we examined these parameters in pregnant rats fed a control (normal weight) or high fat (HF) diet (obese) at 2 critical times of rat placental development. Early trophoblast invasion was increased by approximately 2-fold in HF-fed dams with a concomitant increase in the expression of matrix metalloproteinase 9 protein, a mediator of tissue remodeling and invasion. Furthermore, we observed significantly higher levels of smooth muscle actin surrounding the placental spiral arteries of HF-fed dams, suggesting impaired spiral artery remodeling. Taken together, the results of this study suggest that altered placental development is an important contributor to the poor pregnancy outcomes and increased fetal demise in our model of lifelong maternal obesity.


Asunto(s)
Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Obesidad/patología , Complicaciones del Embarazo/patología , Trofoblastos/patología , Remodelación Vascular/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Longevidad/fisiología , Obesidad/complicaciones , Embarazo , Complicaciones del Embarazo/etiología , Ratas , Ratas Sprague-Dawley
9.
Methods Mol Biol ; 1049: 371-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23913231

RESUMEN

Ovarian cancer is a complex disease, with unclear origins, complicated, multistep tumorigenesis, and variable outcomes. As such, generating experimental models to study the disease and treatment efficacies has proven to be extremely challenging. A number of studies have utilized monolayer in vitro experiments to decipher the cellular changes in ovarian cancer and responses to different treatment approaches. Others have generated three-dimensional spheroid cultures to evaluate cellular function in an environment with more physiological contact with other cells and their matrices. Lastly, a variety of in vivo models have been used to investigate the onset and progression of ovarian cancer and how tumors respond to treatments in an intact physiological environment. This chapter discusses a number of different experimental approaches to study the etiology, biology, and pathology of ovarian tumors and their response to different anticancer therapies.


Asunto(s)
Carcinogénesis , Técnicas de Cultivo de Célula/métodos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Ováricas/etiología , Neoplasias Ováricas/patología
10.
Methods Mol Biol ; 1049: 409-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23913234

RESUMEN

One of the difficulties in studying ovarian cancer historically has been the lack of a suitable animal model that replicates the human disease. Mouse models that utilize intraperitoneal implantation of tumorigenic cells lack interaction between the transformed ovarian epithelial cells and the ovarian stroma, which we have shown to be an integral component in replicating the etiology seen in human epithelial ovarian cancer (Greenaway, Gynecol Oncol 108:385-394, 2008). Xenograft models generally require the use of immunocompromised hosts, which then eliminates the influence of the immune system in disease progression, which also has been shown to be an important part of the progression of epithelial ovarian cancer (EOC). In this chapter, we describe the generation and optimization of an orthotopic, syngeneic mouse model and illustrate the importance of facilitating epithelial-stromal cell interaction to more closely replicate human EOC.


Asunto(s)
Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Células del Estroma/patología , Animales , Carcinoma Epitelial de Ovario , Transformación Celular Neoplásica/genética , Femenino , Humanos , Ratones , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Ovario/patología , Trasplante Heterólogo
11.
PLoS One ; 7(3): e33370, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22442686

RESUMEN

Maternal obesity results in a number of obstetrical and fetal complications with both immediate and long-term consequences. The increased prevalence of obesity has resulted in increasing numbers of women of reproductive age in this high-risk group. Since many of these obese women have been subjected to hypercaloric diets from early childhood we have developed a rodent model of life-long maternal obesity to more clearly understand the mechanisms that contribute to adverse pregnancy outcomes in obese women. Female Sprague Dawley rats were fed a control diet (CON--16% of calories from fat) or high fat diet (HF--45% of calories from fat) from 3 to 19 weeks of age. Prior to pregnancy HF-fed dams exhibited significant increases in body fat, serum leptin and triglycerides. A subset of dams was sacrificed at gestational day 15 to evaluate fetal and placental development. The remaining animals were allowed to deliver normally. HF-fed dams exhibited a more than 3-fold increase in fetal death and decreased neonatal survival. These outcomes were associated with altered vascular development in the placenta, as well as increased hypoxia in the labyrinth. We propose that the altered placental vasculature may result in reduced oxygenation of the fetal tissues contributing to premature demise and poor neonatal survival.


Asunto(s)
Grasas de la Dieta/efectos adversos , Muerte Fetal/fisiopatología , Obesidad/fisiopatología , Placenta/irrigación sanguínea , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Animales , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Femenino , Muerte Fetal/inducido químicamente , Muerte Fetal/metabolismo , Muerte Fetal/patología , Feto/metabolismo , Feto/patología , Feto/fisiopatología , Humanos , Hipoxia/inducido químicamente , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia/fisiopatología , Leptina/sangre , Obesidad/sangre , Obesidad/inducido químicamente , Obesidad/patología , Placenta/patología , Placenta/fisiopatología , Embarazo , Ratas , Ratas Sprague-Dawley , Triglicéridos/sangre
12.
Biol Reprod ; 72(5): 1071-8, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15616224

RESUMEN

Angiogenesis does not normally occur in most adult tissues. However, in the ovary, there are cyclical vascular changes including angiogenesis that involve the interaction of numerous cytokines and growth factors. Angiogenic processes are regulated by a balance between pro- and antiangiogenic factors. The purpose of this study was to determine the expression of the antiangiogenic thrombospondin family and proangiogenic vascular endothelial growth factor (VEGF) in various sizes of healthy bovine follicles. Ovaries were collected from slaughterhouse animals and healthy follicles were sorted based on size (< 0.5 cm, small; 0.5-1.0 cm, medium; >1.0 cm, large). Thrombospondin (TSP) protein levels were significantly higher in small follicles. Immunohistochemistry confirmed the granulosa layer as the primary area within the follicle involved in TSP generation and that small follicles had the highest proportion of immunopositive cells. TSP-1 and -2 mRNA levels were significantly higher in small follicles than either medium or large follicles. TSP colocalized with CD36 on granulosa cells (GC) in the follicle and in cultured cells. In contrast with TSP, VEGF expression increased during growth and development of the follicle. FSH stimulated GC expression of TSP, while LH had no effect. In summary, TSP-1 and -2 were coordinately expressed in the extravascular compartment of the ovary during early follicle development. VEGF was inversely expressed, with expression increasing as follicles developed. Regulated expression and localization of these proteins suggests that they may be involved in regulating growth and development of the follicle in a novel fashion.


Asunto(s)
Folículo Ovárico/metabolismo , Trombospondinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígenos CD36/metabolismo , Bovinos , Femenino , Líquido Folicular/metabolismo , Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Técnicas In Vitro , Neovascularización Fisiológica , Folículo Ovárico/fisiología , Ovario/irrigación sanguínea , Ovario/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Trombospondinas/genética , Factor A de Crecimiento Endotelial Vascular/genética
13.
Biol Reprod ; 67(5): 1522-31, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12390884

RESUMEN

Thrombospondin (TSP)-1 and -2 are extracellular matrix glycoproteins that are both antiangiogenic and important in regulating cellular development, differentiation, and function. To evaluate the expression of TSP in follicular and luteal development, ovarian cycles of Sprague-Dawley rats were synchronized and tissues collected daily at stages corresponding to the early antral, ovulatory, early luteal, and late luteal phases of the ovarian cycle. Immunohistochemistry and Western blot analyses demonstrated that TSP-1 protein and its receptor, CD36, were present in the early antral phase and were localized primarily to the granulosa cells of antral follicles. Both proteins were also present immediately after ovulation and were localized to the developing corpus luteum. Messenger RNA for TSP-1 showed a similar pattern, with expression at the early antral and ovulatory phases. Protein and mRNA expression for TSP-2 was relatively delayed compared to TSP-1, although TSP-2 also was expressed in granulosa cells. Both TSP-1 and -2 were increased in response to LH stimulation in vitro, whereas TSP-2 was suppressed by FSH. The temporal pattern of expression of TSP-1, -2, and CD36, which mirrors the active phases of angiogenesis in this experimental model, is compatible with a role for these proteins in the control of ovarian vascularization.


Asunto(s)
Antígenos CD36/metabolismo , Cuerpo Lúteo/fisiología , Folículo Ovárico/fisiología , Trombospondina 1/metabolismo , Trombospondinas/metabolismo , Animales , Antígenos CD36/genética , Células Cultivadas , Cuerpo Lúteo/citología , Femenino , Hormona Folículo Estimulante/farmacología , Regulación de la Expresión Génica , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/fisiología , Hormona Luteinizante/farmacología , Ciclo Menstrual/fisiología , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Trombospondina 1/efectos de los fármacos , Trombospondina 1/genética , Trombospondinas/efectos de los fármacos , Trombospondinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA