Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Lab Invest ; 103(1): 100017, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36748194

RESUMEN

FoxO1 is an important transcriptional factor that regulates cell survival and metabolism in many tissues. Deleting FoxO1 results in embryonic death due to failure of chorioallantoic fusion at E8.5; however, its role in placental development during mid-late gestation is unclear. In both human patients with gestational diabetes and pregnant mice with hyperglycemia, placental FoxO1 expression was significantly increased. Using FoxO1+/- mice, the effects of FoxO1 haploinsufficiency on placental development under normoglycemia and hyperglycemia were investigated. With FoxO1 haploinsufficiency, the term placental weight increased under both normal and hyperglycemic conditions. Under normoglycemia, this weight change was associated with a general enlargement of the labyrinth, along with increased cell proliferation, decreased cell apoptosis, and decreased expression of p21, p27, Casp3, Casp8, and Rip3. However, under hyperglycemia, the placental weight change was associated with increased fetal blood space, VEGFA overexpression, and expression changes of the angiogenic markers, Eng and Tsp1. In conclusion, FoxO1 plays a role in regulating cell proliferation, cell survival, or angiogenesis, depending on blood glucose levels, during placenta development.


Asunto(s)
Diabetes Gestacional , Proteína Forkhead Box O1 , Hiperglucemia , Animales , Femenino , Humanos , Ratones , Embarazo , Proliferación Celular/genética , Diabetes Gestacional/genética , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Hiperglucemia/genética , Hiperglucemia/metabolismo , Placenta/metabolismo
2.
Lab Invest ; 100(4): 553-569, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31748681

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) has a developmental origin and is influenced in utero. We aimed to evaluate if maternal diet intervention before pregnancy would be beneficial to reduce the risk of offspring NAFLD. In our study, female mice were either on a normal-fat diet (NF group), or a high-fat diet for 12 weeks and continued on this diet throughout pregnancy and lactation (HF group), or switched from HF-to-NF diet 1 week (H1N group), or 9 weeks (H9N group) before pregnancy. Compared with the NF offspring, the H1N and HF, but not the H9N offspring, displayed more severe hepatic steatosis and glucose intolerance. More specifically, an abnormal blood lipid panel was seen in the H1N offspring and abnormal hepatic free fatty acid composition was present in both the HF and H1N offspring, while the H9N offspring displayed both at normal levels. These physiological changes were associated with desensitized hepatic insulin/AKT signaling, increased expression of genes and proteins for de novo lipogenesis and cholesterol synthesis, decreased expression of genes and proteins for fatty acid oxidation, increased Pcsk9 expression, and hypoactivation of 5' AMP-activated protein kinase (AMPK) signaling in the HF and H1N offspring. However, these effects were completely or partially rescued in the H9N offspring. In summary, we found that early maternal diet intervention is effective in reducing the risk of offspring NAFLD caused by maternal HF diet. These findings provide significant support to develop effective diet intervention strategies and policies for prevention of obesity and NAFLD to promote optimal health outcomes for mothers and children.


Asunto(s)
Dieta Alta en Grasa , Metabolismo de los Lípidos , Hígado/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Femenino , Insulina/metabolismo , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Lipogénesis , Masculino , Ratones , Embarazo , Transcriptoma , Aumento de Peso/fisiología
3.
J Nutr Biochem ; 111: 109187, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36270572

RESUMEN

Little is known if and how maternal diet affects the liver phospholipid profiles that contribute to non-alcoholic fatty liver disease (NAFLD) development in offspring. We examined NAFLD phenotypes in male offspring mice of either maternal normal-fat diet (NF group), maternal high-fat diet (HF group), maternal methionine supplement (H1S group), or complete one-carbon supplement (H2S group) added to the maternal HF diet during gestation and lactation. HF offspring displayed worsened NAFLD phenotypes induced by post-weaning HF diet, however, maternal one-carbon supplement prevented such outcome. HF offspring also showed a distinct phospholipid profile from the offspring exposed to H1S or H2S diet. Whole genome bisulfite sequencing (WGBS) analysis further identified five pathways involved in phospholipid metabolism altered by different maternal diet interventions. Furthermore, differential methylated regions (DMRs) on Prkca, Dgkh, Plcb1 and Dgki were identified comparing between HF and NF offspring; most of these DMRs were recovered in H2S offspring. These methylation pattern changes were associated with gene expression changes: HF diet significantly reduced while H1S and H2S diet recovered their levels. Maternal HF diet disrupted offspring phospholipid profiles contributing to worsened hepatic steatosis. The maternal one-carbon supplement prevented such effects, probably through DNA methylation modification.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Efectos Tardíos de la Exposición Prenatal , Femenino , Humanos , Masculino , Ratones , Animales , Dieta Alta en Grasa/efectos adversos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Fosfolípidos/metabolismo , Carbono/metabolismo , Hígado/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo
4.
Toxicol Lett ; 366: 1-6, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35753640

RESUMEN

Cadmium (Cd) is an environmental and occupational pollutant inhaled through smoking or ingested through contaminated food. Yet, little is known about its teratogenicity. In this study, the effects of Cd on embryonic heart development were investigated by exposing Cd to chicken embryos in ovo. Fertilized eggs were treated with Cd at Hamburger-Hamilton Stage (HH)16 and collected at HH35 for histological evaluation of the heart. Cd treatment of 100 µM at HH16 increased embryo mortality at HH35. Specific structural heart defects were not observed in any Cd treatment group, but the relative myocardial tissue area of the right ventricle was increased with Cd exposure. When the HH31 hearts were stained with p-H3S10, the right ventricle had an increased number of cells undergoing proliferation, which was associated with upregulation of Cdk1, Cdk6, CycA, CycD, and CycE detected by qPCR. These findings suggest that Cd exposure from HH16 upregulates proliferation genes and drives overgrowth of the right ventricle. These results grant further attention to Cd teratogenicity on embryonic heart development. Such morphological changes in the heart can potentially affect cardiac function and increase the risk for future cardiovascular diseases, such as heart failure.


Asunto(s)
Cadmio , Miocitos Cardíacos , Animales , Cadmio/toxicidad , Proliferación Celular , Embrión de Pollo , Corazón , Ventrículos Cardíacos , Hiperplasia
5.
Front Biosci (Landmark Ed) ; 26(11): 965-976, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34856745

RESUMEN

Introduction: Hyperglycemic conditions achieved during pregnancy have been shown to have detrimental effects to fetal development and increase the prevalence of childhood comorbidities. However, the mechanisms in which diabetic pregnancies affect placental development and subsequently contribute to adverse health effects on the mother and offspring remain unclear. Research design and methods: Streptozotocin was used to induce gestational diabetes in mice. In this model, hyperglycemia was established at embryonic day 3.5 (E3.5). Pregnancy mass was collected at E10.5, E12.5, E14.5, and E16.5 for different assessments. Results: Both placental and embryonic weights were found to be significantly elevated at E16.5. At E14.5, a significantly larger junctional zone with increased number of glycogen trophoblasts was found in the placentas from hyperglycemic pregnancies (HG group) compared to the placentas from normoglycemic pregnancies (NG group). Importantly, the HG placenta exhibited decreased trophoblast giant cell (TGC) association and TUNEL+ cells, and increased expression of α-SMA on the spiral artery, suggesting arterial remodeling was impacted. Moreover, the interhemal membrane of the labyrinth layer, was found to be thicker in the HG placentas. Furthermore, hyperglycemia resulted in more offspring congenital defects, which were associated with a thicker interhemal membrane. Conclusions: Together, these results suggest that gestational diabetes perturbs proper placental development and function, specifically spiral artery remodeling and angiogenesis, thereby negatively impacting embryonic development.


Asunto(s)
Hiperglucemia , Placenta , Animales , Arterias , Femenino , Ratones , Placentación , Embarazo , Trofoblastos
6.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165955, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877749

RESUMEN

While the correlation between diabetes during pregnancy and birth defects is well-established, how hyperglycemia causes developmental abnormalities remains unclear. In this study, we developed a novel "hyperglycemic" chicken embryonic model by administrating various doses of glucose to fertilized eggs at embryonic stages HH16 or HH24. When the embryos were collected at HH35, the LD50 was 1.57 g/Kg under HH16 treatment and 0.93 g/Kg under HH24 treatment, indicating that "hyperglycemic" environments can be lethal for the embryos. When exposed to a dose equal to or higher than 1 g/Kg glucose at HH16 or HH24, more than 40% of the surviving chicken embryos displayed heart defects and/or limb defects. The limb defects were associated with proliferation defects of both the wing and leg buds indicated by reduced numbers of p-H3S10 labeled cells. These limb defects were also associated with ectopic apoptosis in the leg bud and expression changes of key apoptotic genes. Furthermore, glucose treatment induced decreased expression of genes involved in Shh-signaling, chondrogenesis, and digit patterning in the limb bud. In summary, our data demonstrated that a high-glucose environment induces congenital heart and limb defects associated with disrupted cell proliferation and apoptosis, possibly through depressed Shh-signaling.


Asunto(s)
Apoptosis , Hiperglucemia/patología , Deformidades Congénitas de las Extremidades/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Pollos , Modelos Animales de Enfermedad , Glucosa/administración & dosificación , Glucosa/farmacología , Hiperglucemia/inducido químicamente , Hiperglucemia/genética , Deformidades Congénitas de las Extremidades/inducido químicamente , Deformidades Congénitas de las Extremidades/genética
7.
J Nutr Biochem ; 86: 108495, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32949717

RESUMEN

Novel progress has been made to understand the adverse pathophysiology in the pancreas of offspring exposed to overnutrition in utero. Our study is the first to evaluate whether the adverse effects of maternal overnutrition on offspring ß-cell function are reversible or preventable through preconception maternal diet interventions. Herein, offspring mice were exposed in utero to one of the following: maternal normal-fat diet (NF group), maternal high-fat diet (HF group) or maternal diet transition from an HF to NF diet 9 weeks before pregnancy (H9N group). Offspring mice were subjected to postweaning HF diet for 12 weeks. HF offspring, but not H9N, displayed glucose intolerance and insulin resistance. HF male offspring had enlarged islet ß-cells with reduced ß-cell density, whereas, H9N male offspring did not show these changes. Co-immunofluorescent (Co-IF) staining of glucose transporter 2 (Glut2) and insulin (Ins) revealed significantly more Glut2+Ins- cells, indicative of insulin degranulation, in HF male offspring but not H9N. In addition, Co-IF of insulin and p-H3S10 indicated that ß cells of HF male offspring, but not H9N, had proliferation defects likely due to inhibited protein kinase B (AKT) phosphorylation. In summary, our study demonstrates that maternal H9N diet effectively prevents functional deterioration of ß cells seen in HF male offspring by avoiding ß-cell proliferation defects and degranulation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Células Secretoras de Insulina/patología , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Proliferación Celular , Femenino , Intolerancia a la Glucosa , Homeostasis , Insulina/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/citología , Masculino , Ratones , Obesidad/metabolismo , Páncreas/metabolismo , Fenotipo , Embarazo , Preñez , Efectos Tardíos de la Exposición Prenatal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA