Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Diabetologia ; 63(2): 385-394, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31820038

RESUMEN

AIMS/HYPOTHESIS: This study aimed to determine, in women with gestational diabetes (GDM), the changes in insulin sensitivity (Matsuda Insulin Sensitivity Index; ISOGTT), insulin response and disposition index (DI) from late pregnancy (34-37 weeks gestation, T1), to early postpartum (1-5 days, T2) and late postpartum (6-12 weeks, T3). A secondary aim was to correlate the longitudinal changes in maternal lipids, adipokines, cytokines and weight in relation to the changes in ISOGTT, insulin response and DI. METHODS: ISOGTT, insulin response and DI were calculated at the three time points (T1, T2 and T3) using the results of a 75 g OGTT. Adipokines, cytokines and lipids were measured prior to each OGTT. Linear mixed-effects models were used to compare changes across each time point. Changes in ISOGTT, insulin response and DI were correlated with changes in maternal adipokines, cytokines and lipids at each time point. RESULTS: A total of 27 women completed all assessments. Compared with T1, ISOGTT was 11.20 (95% CI 8.09, 14.31) units higher at 1-5 days postpartum (p < 0.001) and was 5.49 (95% CI 2.38, 8.60) units higher at 6-12 weeks postpartum (p < 0.001). Compared with T1, insulin response values were 699.6 (95% CI 957.5, 441.6) units lower at T2 (p < 0.001) and were 356.3 (95% CI 614.3, 98.3) units lower at T3 (p = 0.004). Compared with T1, the DI was 6434.1 (95% CI 2486.2, 10,381.0) units higher at T2 (p = 0.001) and was 4262.0 (95% CI 314.6, 8209.3) units higher at T3 (p = 0.03). There was a decrease in mean cholesterol, triacylglycerol, LDL-cholesterol and VLDL-cholesterol from T1 to T2 (all p < 0.001), and an increase in mean C-reactive protein, IL-6 and IL-8 from T1 to T2 (all p < 0.001). Mean leptin decreased from T1 to T2 (p = 0.001). There was no significant change in mean adiponectin (p = 0.99) or TNF-α (p = 0.81) from T1 to T2. The mean maternal BMI decreased from T1 to T2 (p = 0.001) and T3 (p < 0.001). There were no significant correlations between any measure of change in ISOGTT, insulin response and DI and change in maternal cytokines, adipokines, lipids or weight from T1 to T2. CONCLUSIONS/INTERPRETATION: In women with GDM, delivery was associated with improvement in both insulin sensitivity and insulin production within the first few days. Improvement in insulin production persisted for 6-12 weeks, but insulin sensitivity deteriorated slightly. These changes in glucose metabolism were not associated to changes in lipids, leptin, inflammation markers or body weight. TRIAL REGISTRATION: ClinicalTrials.gov NCT02082301.


Asunto(s)
Diabetes Gestacional/metabolismo , Periodo Posparto/sangre , Adipoquinas/sangre , Adiponectina/sangre , Adulto , Glucemia/metabolismo , Proteína C-Reactiva/metabolismo , Diabetes Gestacional/sangre , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/sangre , Resistencia a la Insulina/fisiología , Leptina/sangre , Embarazo , Adulto Joven
2.
J Cell Physiol ; 232(12): 3520-3529, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28109119

RESUMEN

Serotonin (5-HT) and its specific transporter, SERT play important roles in pregnancy. Using placentas dissected from 18d gestational SERT-knock out (KO), peripheral 5-HT (TPH1)-KO, and wild-type (WT) mice, we explored the role of 5-HT and SERT in placental functions in detail. An abnormal thick band of fibrosis and necrosis under the giant cell layer in SERT-KO placentas appeared only moderately in TPH1-KO and minimally present in WT placentas. The majority of the changes were located at the junctional zone of the placentas in SERT. The etiology of these findings was tested with TUNEL assays. The placentas from SERT-KO and TPH1-KO showed 49- and 8-fold increase in TUNEL-positive cells without a concurrent change in the DNA repair or cell proliferation compared to WT placentas. While the proliferation rate in the embryos of TPH1-KO mice was 16-fold lower than the rate in gestational age matched embryos of WT or SERT-KO mice. These findings highlight an important role of continuous 5-HT signaling on trophoblast cell viability. SERT may contribute to protecting trophoblast cells against cell death via terminating the 5-HT signaling which changes cell death ratio in trophoblast as well as proliferation rate in embryos. However, the cell death in SERT-KO placentas is in caspase 3-independent pathway.


Asunto(s)
Apoptosis , Caspasa 3/metabolismo , Placenta/enzimología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Animales , Glucemia/metabolismo , Proliferación Celular , Femenino , Genotipo , Insulina/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Placenta/metabolismo , Embarazo , Serotonina/sangre , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Transducción de Señal , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
3.
Diabetologia ; 59(11): 2459-2466, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27535280

RESUMEN

AIMS/HYPOTHESIS: Obesity triggers complex inflammatory networks within the innate immune system. During pregnancy, the placenta amplifies the low-grade inflammation through activation of Toll-like receptor 4 (TLR4) signalling pathways. The purpose of this study was to investigate the impact of obesity on placental TLR4 expression and inflammatory signals. The secondary aim was to analyse the placental cell type responsible for TLR4 activation. METHODS: Thirty-nine women recruited at term-scheduled Caesarean section were grouped according to their pre-gravid BMI (<25 kg/m(2) and >30 kg/m(2)). Placenta, venous maternal and cord blood were obtained at delivery for analysis. Data were analysed with linear regression and Spearman's rank correlation coefficient analysis. RESULTS: TLR4, IL6 and IL8 expression was increased three- to ninefold (p < 0.001) in the placenta of obese vs lean women. There was a positive correlation between placental TLR4 and maternal systemic and placental IL6 and IL8 concentrations. Placental TLR4 expression was correlated with maternal pre-gravid BMI, insulin resistance index, plasma insulin and C-reactive protein (r = 0.57, 0.31, 0.35, 0.53, respectively; p < 0.001) but not with plasma glucose, maternal age, gestational age and gestational weight gain (r < 0.2; p > 0.1). TLR4 was located in both trophoblast and macrovascular endothelial cells lining fetal vasculature. Lipopolysaccharide-induced TLR4 activation was more robust in trophoblasts than in endothelial vascular cells (100-fold vs tenfold; p < 0.001). CONCLUSIONS/INTERPRETATION: Trophoblastic TLR4 is strongly implicated in the propagation of placental inflammation. Placental inflammation is related to maternal metabolic conditions such as pre-gravid BMI, whilst gestational weight gain or gestational age are not. These results implicate the pre-gravid condition as a significant contributor to metabolic inflammation in late pregnancy.


Asunto(s)
Placenta/metabolismo , Receptor Toll-Like 4/metabolismo , Proteína C-Reactiva/metabolismo , Femenino , Edad Gestacional , Humanos , Inflamación/sangre , Inflamación/metabolismo , Insulina/sangre , Resistencia a la Insulina , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Obesidad , Embarazo , Transducción de Señal/fisiología
4.
Am J Obstet Gynecol ; 214(6): 745.e1-5, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26719212

RESUMEN

BACKGROUND: Weight gain in pregnancy is an essential physiologic adaptation that supports growth and development of a fetus and is distributed among lean mass that includes total body water and fat mass gains. Although gestational weight gain provides a source of energy for the mother and fetus, excess gestational weight gain may underlie reported associations between parity and future metabolic disorders and is linked to postpartum weight retention and insulin resistance. Although weight gain often is proposed as a modifiable variable to mitigate adverse maternal and offspring health outcomes, our knowledge of specific maternal body composition changes with weight gain and the potential metabolic consequences is limited. Furthermore, although gestational weight gain alters maternal body composition, the impact of excess weight gain on fat and lean mass is not well-studied. Understanding the accrual of fat and lean body mass may improve our understanding of the role of excessive gestational weight gain and metabolic dysfunction. OBJECTIVE: The purpose of our study was to quantify the relationship between gestational weight gain and maternal fat and lean body mass accrual and to compare fat and lean body mass accrual according to the 2009 Institute of Medicine Guidelines for Gestational Weight Gain in Pregnancy adherence. We hypothesized that exceeding current weight gain guidelines would be associated with greater fat, compared with lean body, mass accrual. STUDY DESIGN: This is a secondary analysis of a randomized controlled trial of 49 overweight/obese women; all 49 are included in this secondary analysis. Maternal weight and body composition were collected in early (13 0/6 to 16 6/7 weeks gestation) and late (34 0/7 to 36 6/7 weeks gestation) pregnancy with the use of air densitometry. Correlations were drawn between gestational weight gain and change in fat and lean body mass. We compared change in fat and lean body mass by adherence to the 2009 Institute of Medicine Guidelines for Gestational Weight Gain in Pregnancy. Nonparametric tests and chi-square analyses were performed; a probability value of <.05 was significant. RESULTS: Early pregnancy body mass index was 30.3 kg/m(2) (interquartile range [IQR], 28.5-35.2 kg/m(2)); women gained 9.0 kg (IQR, 5.3-13.2 kg). Overweight and obese women were equally likely to gain excess weight (48% vs 35%; P = .6). Weight gain correlated strongly with fat mass change (r = 0.87; P < .001); women with excess vs adequate vs inadequate weight gain had greater fat mass change overall (5.2 [IQR, 4.2-8.1] vs 0.2 [IQR, -0.4-2.2] vs -2.7 [IQR, -5.2- -0.7] kg, respectively; P < .001) and in all pairwise comparisons. Weight gain also correlated with lean body mass change (r = 0.52; P = .001), but women with excess vs adequate weight gain had similar lean body mass change (8.4 [IQR, 7.2-10.1] vs 7.8 [IQR, 6.0-8.7] kg; P = .1). CONCLUSION: Excess gestational weight gain is associated primarily with maternal fat, but not with lean body mass accrual. Our results may help explain the reason that excess gestational weight gain or fat mass accrual is associated with long-term obesity, metabolic dysfunction, and cardiovascular disease risk.


Asunto(s)
Distribución de la Grasa Corporal , Obesidad/fisiopatología , Sobrepeso/fisiopatología , Aumento de Peso/fisiología , Adulto , Índice de Masa Corporal , Estudios de Cohortes , Femenino , Humanos , Embarazo , Adulto Joven
5.
Diabetologia ; 58(7): 1561-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25957777

RESUMEN

AIMS/HYPOTHESIS: Maternal metabolic alterations are essential to achieve healthy pregnancy outcomes, but increasing maternal parity may be associated with long-term metabolic dysfunction risk. As existing data are limited by study design, our aim was to employ robust metabolic measures to determine whether or not physiological pregnancy alterations in maternal metabolic function persist at 1 year postpartum. METHODS: We evaluated 21 healthy women, of whom 11 had an interval pregnancy (IP) and assessment at preconception, during pregnancy and 1 year postpartum, and 10 had no IP and assessment at baseline and a 1 year interval. Assessment measures included body composition, insulin sensitivity and response, and basal metabolic rate. For each measure, IP vs no IP and time intervals within each group were compared using nonparametric analyses, reporting median (IQR). RESULTS: IP and no IP women were similar at enrolment, and no IP women had similar metabolic profiles at enrolment and the 1 year interval. IP women exhibited expected metabolic changes during pregnancy compared with preconception. In IP women, preconception and postpartum measures, including fat mass (20.7 [13.7-37.4] kg vs 18.4 [13.8-41.3] kg; p = 0.2), total insulin response (AUC 11,459 [9,230-13,696] pmol/ml × min vs 11,522 [5,882-17,404] pmol/ml × min; p = 0.9), insulin sensitivity (0.12 [0.06-0.13] mg [kg fat-free mass (FFM)](-1) min(-1) vs 0.11 [0.10-0.15] mg [kg FFM](-1) min(-1); p = 0.1) and basal metabolic rate (0.092 [0.092-0.105] kJ min(-1) FFM vs 0.096 [0.088-0.096] kJ min(-1) FFM; p = 0.5), were similar. CONCLUSIONS/INTERPRETATION: Our findings suggest pregnancy might not irreversibly alter maternal metabolic profile, measured at preconception through to 1 year postpartum. This result might be explained by a return to pre-pregnancy weight.


Asunto(s)
Periodo Posparto/metabolismo , Embarazo/metabolismo , Adulto , Metabolismo Basal , Composición Corporal , Distribución de la Grasa Corporal , Estatura , Calorimetría Indirecta , Diabetes Mellitus/genética , Escolaridad , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Estudios Longitudinales , Enfermedades Metabólicas/epidemiología , Enfermedades Metabólicas/metabolismo , Paridad , Estudios Prospectivos
6.
Hum Reprod ; 30(9): 2152-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26202921

RESUMEN

STUDY QUESTION: What are the effects of fatty acids on placental inflammatory cytokine with respect to toll-like receptor-4/nuclear factor-kappa B (TLR4/NF-kB)? SUMMARY ANSWER: Exogenous fatty acids induce a pro-inflammatory cytokine response in human placental cells in vitro via activation of TLR4 signaling pathways. WHAT IS KNOWN ALREADY: The placenta is exposed to changes in circulating maternal fatty acid concentrations throughout pregnancy. Fatty acids are master regulators of innate immune pathways through recruitment of toll-like receptors and activation of cytokine synthesis. STUDY DESIGN, SIZE, DURATION: Trophoblast cells isolated from 14 normal term human placentas were incubated with long chain fatty acids (FA) of different carbon length and degree of saturation. The expression and secretion of interleukin-6 (IL-6), IL-8 and tumor necrosis factor-alpha (TNF-α) were measured by reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Antibodies against TLR4 ligand binding domain, downstream signaling and anti-p65 NFkB-inhibitor were used to characterize the pathways of FA action. PARTICIPANTS/MATERIALS, SETTING, METHODS: General approach used primary human term trophoblast cell culture. Methods and end-points used real-time quantitative PCR, cytokine measurements, immunohistochemistry, western blots. MAIN RESULTS AND THE ROLE OF CHANCE: The long chain saturated fatty acids, stearic and palmitic (PA), stimulated the synthesis as well as the release of TNF-α, IL-6 and IL-8 by trophoblast cells (2- to 6-fold, P < 0.001). In contrast, the unsaturated (palmitoleic, oleic, linoleic) acids did not modify cytokine expression significantly. Palmitate-induced inflammatory effects were mediated via TLR4 activation, NF-kB phosphorylation and nuclear translocation. LIMITATIONS, REASONS FOR CAUTION: TNF-α protein level was close to the limit of detection in the culture medium even when cells were cultured with PA. WIDER IMPLICATIONS OF THE FINDINGS: These mechanisms open the way to a better understanding of how changes in maternal lipid homeostasis may regulate placental inflammatory status. STUDY FUNDING/COMPETING INTERESTS: X.Y. was recipient of fellowship award from West China Second University Hospital, Sichuan University (NIH HD 22965-19). The authors have nothing else to disclose. TRIAL REGISTRATION NUMBER: None.


Asunto(s)
Citocinas/metabolismo , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Trofoblastos/metabolismo , Adulto , Femenino , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , FN-kappa B/metabolismo , Embarazo , Trofoblastos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
7.
Am J Obstet Gynecol ; 212(5): 647.e1-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25731694

RESUMEN

OBJECTIVE: The purpose of this study was to investigate the effects of insulin on human placental transcriptome and biological processes in first-trimester pregnancy. STUDY DESIGN: Maternal plasma and placenta villous tissue were obtained at the time of voluntary termination of pregnancy (7-12 weeks) from 17 lean (body mass index, 20.9±1.5 kg/m2) and 18 obese (body mass index, 33.5±2.6 kg/m2) women. Trophoblast cells were immediately isolated for in vitro treatment with insulin or vehicle. Patterns of global gene expression were analyzed using genome microarray profiling after hybridization to Human Gene 1.1 ST and real time reverse transcription-polymerase chain reaction. RESULTS: The global trophoblast transcriptome was qualitatively separated in insulin-treated vs untreated trophoblasts of lean women. The number of insulin-sensitive genes detected in the trophoblasts of lean women was 2875 (P<.001). Maternal obesity reduced the number of insulin-sensitive genes recovered by 30-fold. Insulin significantly impaired several gene networks regulating cell cycle and cholesterol homeostasis but did not modify pathways related to glucose transport. Obesity associated with high insulin and insulin resistance, but not maternal hyperinsulinemia alone, impaired the global gene profiling of early gestation placenta, highlighting mitochondrial dysfunction and decreased energy metabolism. CONCLUSION: We report for the first time that human trophoblast cells are highly sensitive to insulin regulation in early gestation. Maternal obesity associated with insulin resistance programs the placental transcriptome toward refractoriness to insulin with potential adverse consequences for placental structure and function.


Asunto(s)
Hipoglucemiantes/farmacología , Insulina/farmacología , Obesidad/genética , Placenta/efectos de los fármacos , Complicaciones del Embarazo/genética , ARN Mensajero/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Trofoblastos/efectos de los fármacos , Aborto Inducido , Adolescente , Adulto , Estudios de Casos y Controles , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Humanos , Obesidad/metabolismo , Placenta/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , Primer Trimestre del Embarazo , ARN Mensajero/metabolismo , Transcriptoma/genética , Trofoblastos/metabolismo , Adulto Joven
8.
Am J Obstet Gynecol ; 209(2): 116.e1-5, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23583837

RESUMEN

OBJECTIVE: The dawn phenomenon is a transient rise in blood glucose between 4 and 6 am that is attributed to the pulsatile release of pituitary growth hormone (GH). In pregnancy, GH is suppressed by placental GH. Hence, we hypothesize that there is no evidence for the dawn phenomenon in late pregnancy in healthy women. STUDY DESIGN: Twenty glucose-tolerant women with singleton gestations between 28 weeks and 36 weeks 6 days' gestation were recruited. The women were admitted overnight to the Clinical Research Unit and had continuous glucose monitoring. Insulin and GH were measured at 2-hour intervals from 8 pm to 8 am. GH was grouped into times 1A (8-10 pm), 2A (12-2 am), and 3A (4-8 am) for changes over time. Further analysis was performed with time 1B (8 pm to 2 am) and 2B (4-8 am). Insulin was measured between 4 and 8 am. RESULTS: Plasma glucose decreased over time (P < .001). There were no significant changes in GH among times 1A, 2A, and 3A (P = .45) or times 1B and 2B (P = .12). Insulin concentrations increased after meals, but there were no changes from 4 am (8.5 ± 1.4 µU/mL) through 8 am (8.6 ± 1.1 µU/mL; P = .98). CONCLUSION: Glucose and insulin concentrations show no increase from 4-8 am; although there is variability in GH, there is no evidence for the dawn phenomenon in late pregnancy in healthy women.


Asunto(s)
Glucemia/análisis , Ritmo Circadiano , Hormona de Crecimiento Humana/sangre , Embarazo/sangre , Adulto , Femenino , Humanos , Insulina/sangre , Factor de Crecimiento Placentario , Proteínas Gestacionales/fisiología
9.
Am J Physiol Endocrinol Metab ; 303(7): E832-40, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22811467

RESUMEN

Changes in adipose tissue metabolism are central to adaptation of whole body energy homeostasis to pregnancy. To gain insight into the molecular mechanisms supporting tissue remodeling, we have characterized the longitudinal changes of the adipose transcriptome in human pregnancy. Healthy nonobese women recruited pregravid were followed in early (8-12 wk) and in late (36-38 wk) pregnancy. Adipose tissue biopsies were obtained in the fasting state from the gluteal depot. The adipose transcriptome was examined via whole genome DNA microarray. Expression of immune-related genes and extracellular matrix components was measured using real-time RT-PCR. Adipose mass, adipocyte size, and cell number increased in late pregnancy compared with pregravid measurements (P < 0.001) but remained unchanged in early pregnancy. The adipose transcriptome evolved during pregnancy with 10-15% of genes being differently expressed compared with pregravid. Functional gene cluster analysis revealed that the early molecular changes affected immune responses, angiogenesis, matrix remodeling, and lipid biosynthesis. Increased expression of macrophage markers (CD68, CD14, and the mannose-6 phosphate receptor) emphasized the recruitment of the immune network in both early and late pregnancy. The TLR4/NF-κB signaling pathway was enhanced specifically in relation to inflammatory adipokines and chemokines genes. We conclude that early recruitment of metabolic and immune molecular networks precedes the appearance of pregnancy-related physiological changes in adipose tissue. This biphasic pattern suggests that physiological inflammation is an early step preceding the development of insulin resistance, which peaks in late pregnancy.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo/fisiología , Inflamación/fisiopatología , Primer Trimestre del Embarazo/fisiología , Tercer Trimestre del Embarazo/fisiología , Adipoquinas/genética , Adipoquinas/inmunología , Adipoquinas/metabolismo , Tejido Adiposo/inmunología , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Antígenos de Diferenciación Mielomonocítica/inmunología , Quimiocinas/genética , Quimiocinas/inmunología , Quimiocinas/metabolismo , Femenino , Humanos , Inflamación/genética , Inflamación/inmunología , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/inmunología , Metabolismo de los Lípidos/fisiología , Receptores de Lipopolisacáridos/biosíntesis , Receptores de Lipopolisacáridos/inmunología , FN-kappa B/inmunología , FN-kappa B/metabolismo , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Embarazo , Primer Trimestre del Embarazo/genética , Primer Trimestre del Embarazo/inmunología , Tercer Trimestre del Embarazo/genética , Tercer Trimestre del Embarazo/inmunología , Receptor IGF Tipo 2/biosíntesis , Receptor IGF Tipo 2/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Transcriptoma/genética , Transcriptoma/inmunología , Transcriptoma/fisiología
10.
J Clin Endocrinol Metab ; 107(9): e3759-e3768, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35686573

RESUMEN

CONTEXT: Pregnancy is characterized by progressive decreases in glucose insulin sensitivity. Low insulin sensitivity resulting in hyperglycemia is associated with higher neonatal adiposity. However, less is known regarding lipid metabolism, particularly lipid insulin sensitivity in pregnancy and neonatal adiposity. OBJECTIVE: Because higher maternal prepregnancy body mass index is strongly associated with both hyperlipidemia and neonatal adiposity, we aimed to examine the longitudinal changes in basal and clamp maternal lipid metabolism as contributors to neonatal adiposity. METHODS: Twelve women planning a pregnancy were evaluated before pregnancy, in early (12-14 weeks), and late (34-36 weeks) gestation. Body composition was estimated using hydrodensitometry. Basal and hyperinsulinemic-euglycemic clamp glucose and glycerol turnover (GLYTO) were measured using 2H2-glucose and 2H5-glycerol and substrate oxidative/nonoxidative metabolism with indirect calorimetry. Total body electrical conductivity was used to estimate neonatal body composition. RESULTS: Basal free-fatty acids decreased with advancing gestation (P = 0.0210); however, basal GLYTO and nonoxidative lipid metabolism increased over time (P = 0.0046 and P = 0.0052, respectively). Further, clamp GLYTO and lipid oxidation increased longitudinally over time (P = 0.0004 and P = 0.0238, respectively). There was a median 50% increase and significant positive correlation during both basal and clamp GLYTO from prepregnancy through late gestation. Neonatal adiposity correlated with late pregnancy basal and clamp GLYTO (r = 0.6515, P = 0.0217; and r = 0.6051, P = 0.0371). CONCLUSIONS: Maternal prepregnancy and late pregnancy measures of basal and clamp lipid metabolism are highly correlated. Late pregnancy basal and clamp GLYTO are significantly associated with neonatal adiposity and account for ~40% of the variance in neonatal adiposity. These data emphasize the importance of maternal lipid metabolism relating to fetal fat accrual.


Asunto(s)
Adiposidad , Resistencia a la Insulina , Ácidos Grasos no Esterificados , Femenino , Glucosa/metabolismo , Glicerol , Humanos , Recién Nacido , Insulina/metabolismo , Metabolismo de los Lípidos , Estudios Longitudinales , Obesidad/metabolismo , Embarazo
11.
Am J Obstet Gynecol ; 204(6): 479-87, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21288502

RESUMEN

The Pedersen hypothesis was formulated more than 50 years ago. Jorgen Pedersen primarily cared for women with type 1 diabetes. He suggested that fetal overgrowth was related to increased transplacental transfer of glucose, stimulating the release of insulin by the fetal beta cell and subsequent macrosomia. Optimal maternal glucose control decreased perinatal mortality and morbidity. However, over the ensuing decades, there have been increases in maternal obesity and subsequently gestational diabetes mellitus (GDM) and type 2 diabetes. The underlying pathophysiology of type 1 and GDM/type 2 diabetes are fundamentally different, type 1 diabetes being primarily a disorder of beta cell failure and type 2 diabetes/GDM including both insulin resistance and beta cell dysfunction. As such the metabolic milieu in which the developing fetus is exposed may be quite different in type 1 diabetes and obesity. In this review we examine the metabolic environment of obese diabetic women and lipid metabolism affecting fetal adiposity. The importance of understanding these issues relates to the increasing trends of obesity worldwide with perinatal programming of metabolic dysfunction in the offspring.


Asunto(s)
Diabetes Gestacional/metabolismo , Macrosomía Fetal/etiología , Feto/metabolismo , Obesidad/epidemiología , Obesidad/metabolismo , Embarazo en Diabéticas/metabolismo , Femenino , Homeostasis , Humanos , Inflamación/metabolismo , Metabolismo de los Lípidos , Embarazo
12.
Am J Obstet Gynecol ; 205(3): 265.e1-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22071058

RESUMEN

OBJECTIVE: The purpose of this study was to gain insight into the pathways that are associated with inflammation at the maternal-fetal interface. This study examined the molecular characteristics of monocytes that were derived from the maternal circulation and the placenta of obese women. STUDY DESIGN: Mononuclear cells were isolated from placenta, venous maternal, and umbilical cord blood at term delivery; activated monocytes were separated with CD14 immunoselection. The genotype and expression pattern of the monocytes were analyzed by microarray and real-time reverse transcriptase-polymerase chain reaction. RESULTS: The transcriptome of the maternal blood and placental CD14 monocytes exhibited 73% homology, with 10% (1800 common genes) differentially expressed. Genes for immune sensing and regulation, matrix remodeling, and lipid metabolism were enhanced 2-2006 fold in placenta, compared with maternal monocytes. The CD14 placental monocytes exhibited a maternal genotype (9% DYS14 expression) as opposed to the fetal genotype (90% DYS14 expression) of the trophoblast cells. CONCLUSION: CD14 monocytes from the maternal blood and the placenta share strong phenotypic and genotypic similarities with an enhanced inflammatory pattern in the placenta. The functional traits of the CD14 blood and placental monocytes suggest that they both contribute to propagation of inflammation at the maternal-fetal interface.


Asunto(s)
Sangre Fetal/metabolismo , Feto/metabolismo , Inflamación/metabolismo , Monocitos/metabolismo , Placenta/metabolismo , Adulto , Femenino , Sangre Fetal/inmunología , Feto/inmunología , Expresión Génica , Humanos , Inflamación/inmunología , Receptores de Lipopolisacáridos/metabolismo , Monocitos/inmunología , Obesidad/inmunología , Obesidad/metabolismo , Placenta/inmunología , Embarazo , Transcriptoma
13.
Nutrients ; 13(2)2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572368

RESUMEN

Maternal obesity, a state of chronic low-grade metabolic inflammation, is a growing health burden associated with offspring adiposity, abnormal fetal growth and prematurity, which are all linked to adverse offspring cardiometabolic health. Higher intake of anti-inflammatory omega-3 (n-3) polyunsaturated fatty acids (PUFA) in pregnancy has been associated with lower adiposity, higher birthweight and longer gestation. However, the effects of n-3 supplementation specifically in pregnant women with overweight and obesity (OWOB) have not been explored. We conducted a pilot double-blind randomized controlled trial of 72 pregnant women with first trimester body mass index (BMI) ≥ 25 kg/m2 to explore preliminary efficacy of n-3 supplementation. Participants were randomized to daily DHA plus EPA (2 g/d) or placebo (wheat germ oil) from 10-16 weeks gestation to delivery. Neonatal body composition, fetal growth and length of gestation were assessed. For the 48 dyads with outcome data, median (IQR) maternal BMI was 30.2 (28.2, 35.4) kg/m2. In sex-adjusted analyses, n-3 supplementation was associated with higher neonatal fat-free mass (ß: 218 g; 95% CI 49, 387) but not with % body fat or fat mass. Birthweight for gestational age z-score (-0.17 ± 0.67 vs. -0.61 ± 0.61 SD unit, p = 0.02) was higher, and gestation longer (40 (38.5, 40.1) vs. 39 (38, 39.4) weeks, p = 0.02), in the treatment vs. placebo group. Supplementation with n-3 PUFA in women with OWOB led to higher lean mass accrual at birth as well as improved fetal growth and longer gestation. Larger well-powered trials of n-3 PUFA supplementation specifically in pregnant women with OWOB should be conducted to confirm these findings and explore the long-term impact on offspring obesity and cardiometabolic health.


Asunto(s)
Composición Corporal/efectos de los fármacos , Ácidos Grasos Omega-3/administración & dosificación , Desarrollo Fetal/efectos de los fármacos , Edad Gestacional , Obesidad/complicaciones , Complicaciones del Embarazo/tratamiento farmacológico , Adiposidad/efectos de los fármacos , Adulto , Antiinflamatorios/administración & dosificación , Peso al Nacer , Suplementos Dietéticos , Método Doble Ciego , Femenino , Humanos , Recién Nacido , Masculino , Obesidad/tratamiento farmacológico , Proyectos Piloto , Embarazo
14.
Clin Sci (Lond) ; 119(3): 123-9, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20443782

RESUMEN

Increasing maternal obesity is a challenge that has an impact on all aspects of female reproduction. Lean and obese pregnant women gain similar fat mass, but lean women store fat in the lower-body compartment and obese women in central compartments. In the non-pregnant, central storage of fat is associated with adipocyte hypertrophy and represents a failure to adequately store excess fatty acids, resulting in metabolic dysregulation and ectopic fat accumulation (lipotoxicity). Obese pregnancy is associated with exaggerated metabolic adaptation, endothelial dysfunction and increased risk of adverse pregnancy outcome. We hypothesize that the preferential storage of fat in central rather than 'safer' lower-body depots in obese pregnancy leads to lipotoxicity. The combination of excess fatty acids and oxidative stress leads to the production of oxidized lipids, which can be cytotoxic and influence gene expression by acting as ligands for nuclear receptors. Lipid excess and oxidative stress provoke endothelial dysfunction. Oxidized lipids can inhibit trophoblast invasion and influence placental development, lipid metabolism and transport and can also affect fetal developmental pathways. As lipotoxicity has the capability of influencing both maternal endothelial function and placental function, it may link maternal obesity and placentally related adverse pregnancy outcomes such as miscarriage and pre-eclampsia. The combination of excess/altered lipid nutrient supply, suboptimal in utero metabolic environment and alterations in placental gene expression, inflammation and metabolism may also induce obesity in the offspring.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Obesidad/metabolismo , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal , Distribución de la Grasa Corporal , Endotelio Vascular/fisiopatología , Femenino , Humanos , Recién Nacido , Obesidad/etiología , Obesidad/fisiopatología , Estrés Oxidativo/fisiología , Embarazo , Complicaciones del Embarazo/fisiopatología , Resultado del Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal/fisiología
15.
Am J Obstet Gynecol ; 201(2): 209.e1-209.e10, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19560108

RESUMEN

OBJECTIVE: Changes in metabolic homeostasis in pregnant diabetic women are potential determinants of increased adiposity of the fetus. The aim of this study was to characterize diabetes mellitus-induced changes in genes for fetoplacental energy metabolism in relation to fetal adiposity. STUDY DESIGN: Placentas of women with type 1 diabetes mellitus, gestational diabetes mellitus (GDM), or no complications were analyzed by microarray profiling. The pattern of gene expression was assessed in primary placental cell cultures. RESULTS: Diabetes mellitus was associated with 49 alterations in gene expression at key steps in placental energy metabolism, with 67% of the alterations related to lipid pathways and 9% of the alterations related to glucose pathways. Preferential activation of lipid genes was observed in pregnancy with GDM. Type 1 diabetes mellitus induced fewer lipid modifications but an enhancement of glycosylation and acylation pathways. Oleate enhanced expression of genes for fatty acid esterification and the formation of lipid droplets 3 times as much as glucose in cultured placental cells. CONCLUSION: These results point to fatty acids as preferential lipogenic substrates for placental cells and suggest that genes for fetoplacental lipid metabolism are enhanced selectively in GDM. The recruited genes may be instrumental in increasing transplacental lipid fluxes and the delivery of lipid substrates for fetal use.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Perfilación de la Expresión Génica , Metabolismo de los Lípidos/genética , Tejido Adiposo/fisiología , Adulto , Glucemia/genética , Glucemia/metabolismo , Células Cultivadas , Metabolismo Energético/genética , Femenino , Feto/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Homeostasis/genética , Humanos , Obesidad/genética , Obesidad/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Placenta/citología , Placenta/fisiología , Embarazo
16.
Mol Aspects Med ; 66: 21-30, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30513311

RESUMEN

Early pregnancy is characterized by a series of complex and tightly regulated events to ultimately establish implantation and early placental development. One of the key events is the opening of the decidual spiral arteries into the intervillous space. It leads to a rise in oxygen tension in the intervillous space and the placenta and will induce transcriptional and translational changes of oxygen-sensitive molecules including antioxidants. Diabetes and/or obesity ('diabesity') are associated with changes in the maternal environment, which can affect any of the distinct developmental processes ensuing modifications of onset or magnitude of oxygen tension changes. This may overwhelm the anti-oxidative defence systems developing in parallel to the physiological rise in oxygen tension. The resulting exacerbated oxidative stress, as it was demonstrated in the first trimester placentas of type 1 diabetes mellitus (T1DM) patients, may impair developmental processes. In addition, many components of the diabesity environment can have distinct molecular effects on a range of molecules, but these need to be identified. Insulin is an important contributor to early placental phenotype, because it is involved in regulation of cytotrophoblast-syncytiotrophoblast fusion and placental surface expansion. Its circulating levels are increased in T1DM, because of pharmacologic treatment, and obesity, because of beta-cell compensation of insulin resistance. This constitutes the (patho)physiological link between diabesity and placental growth changes. Microarray studies have identified several molecular and cellular candidate processes altered by insulin in obese pregnancies, including cell cycle regulation and fatty acid and cholesterol metabolism. Research on early diabesity exposure and the placenta is still in its infant stage. To stimulate further studies we have identified some important and pending questions.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Placenta/metabolismo , Femenino , Humanos , Estrés Oxidativo , Placentación , Embarazo , Transducción de Señal
18.
Endocrinology ; 158(8): 2543-2555, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28541534

RESUMEN

Obese women, on average, give birth to babies with high fat mass. Placental lipid metabolism alters fetal lipid delivery, potentially moderating neonatal adiposity, yet how it is affected by maternal obesity is poorly understood. We hypothesized that fatty acid (FA) accumulation (esterification) is higher and FA ß-oxidation (FAO) is lower in placentas from obese, compared with lean women. We assessed acylcarnitine profiles (lipid oxidation intermediates) in mother-baby-placenta triads, in addition to lipid content, and messenger RNA (mRNA)/protein expression of key regulators of FA metabolism pathways in placentas of lean and obese women with normal glucose tolerance recruited at scheduled term Cesarean delivery. In isolated trophoblasts, we measured [3H]-palmitate metabolism. Placentas of obese women had 17.5% (95% confidence interval: 6.1, 28.7%) more lipid than placentas of lean women, and higher mRNA and protein expression of FA esterification regulators (e.g., peroxisome proliferator-activated receptor γ, acetyl-CoA carboxylase, steroyl-CoA desaturase 1, and diacylglycerol O-acyltransferase-1). [3H]-palmitate esterification rates were increased in trophoblasts from obese compared with lean women. Placentas of obese women had fewer mitochondria and a lower concentration of acylcarnitines, suggesting a decrease in mitochondrial FAO capacity. Conversely, peroxisomal FAO was greater in placentas of obese women. Altogether, these changes in placental lipid metabolism may serve to limit the amount of maternal lipid transferred to the fetus, restraining excess fetal adiposity in this population of glucose-tolerant women.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Obesidad/metabolismo , Placenta/metabolismo , Adulto , Peso Corporal , Ácidos Grasos , Femenino , Regulación de la Expresión Génica , Humanos , Recién Nacido , Masculino , Embarazo , Adulto Joven
19.
J Clin Endocrinol Metab ; 91(1): 248-55, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16249288

RESUMEN

CONTEXT: Obesity and diabetes during pregnancy are associated with increased insulin resistance and higher neonatal adiposity. In turn, insulin resistance triggers inflammatory pathways with accumulation of placental cytokines. OBJECTIVE: To determine placental signals that translate into development of excess adipose tissue, we investigated the role of phospholipases A2 (PLA2) as targets of inflammatory mediators. SETTING: The study was conducted at Case Western Reserve University, Department of Reproductive Biology. SUBJECTS: Volunteers gave informed written consent in accordance with the Institutional Review Board guidelines. Placenta and cord blood samples were obtained at the time of elective cesarean section in 15 term pregnancies. INTERVENTION: Neonatal anthropometric measurements were performed within 48 h of delivery. Placentas were grouped based on neonatal percentage body fat as obese (body fat > or = 16%) and lean control (body fat < or = 8%). MAIN OUTCOME MEASURES: The primary outcomes were placenta PLA2 expression and fatty acid concentration. RESULTS: Expression of PLA2G2A and PLA2G5, the main placenta phospholipases, was greater (P < 0.05) in placenta of obese compared with control neonates and was associated with increased 20:3 and 20:5 omega-3 polyunsaturated fatty acids. TNF-alpha and leptin content was increased 3-fold in placenta of obese neonates. TNF-alpha and leptin both induced a time-dependent activation of PLA2G2 and PLA2G5 in placental cells. CONCLUSION: Accumulation of omega-3 fatty acids through secretory PLA2 activation is associated with high neonatal adiposity. We propose that the generation of placental lipid mediators through TNF-alpha and leptin stimulation represents a key mechanism to favor excess fetal fat accretion.


Asunto(s)
Enfermedades Fetales/enzimología , Enfermedades Fetales/fisiopatología , Lípidos/fisiología , Obesidad/enzimología , Obesidad/fisiopatología , Fosfolipasas A/metabolismo , Placenta/fisiología , Adulto , Separación Celular , Células Cultivadas , Activación Enzimática , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Femenino , Humanos , Recién Nacido , Leptina/sangre , Fosfolipasas A2 , Placenta/citología , Placenta/enzimología , Embarazo , ARN/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
20.
Am J Obstet Gynecol ; 194(6): 1537-45, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16731069

RESUMEN

Leptin, which was identified originally as an adipocyte-derived protein, was regarded for years as an exclusive regulator of satiety and energy homeostasis. A role for leptin in pregnancy was later suggested by the findings that plasma levels during gestation are greater than in nongravid individuals and that leptin is synthesized within the fetoplacental unit. Observational studies have established that leptin production is dysregulated in several pathologic stages of pregnancy in association with alterations of fetal growth. For example, an overproduction of leptin by the placenta in pregnancy with diabetes mellitus or hypertension is associated with maternal hyperleptinemia. Evidence is also accumulating that umbilical leptin levels can be viewed as a biomarker of fetal adiposity. Ten years after its discovery as a hormone, we review the known and unknowns of leptin in pregnancy with particular emphasis on its functions in health and disease. We aim to demonstrate that studies of leptin in pregnancy largely have contributed to insight into the mechanisms of leptin action, both as a hormone and as a cytokine.


Asunto(s)
Leptina/fisiología , Embarazo/fisiología , Animales , Femenino , Sangre Fetal , Desarrollo Fetal , Feto/fisiología , Humanos , Leptina/sangre , Placenta/metabolismo , Placenta/fisiología , Embarazo/sangre , Complicaciones del Embarazo/sangre , Receptores de Superficie Celular/metabolismo , Receptores de Leptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA