Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Commun Med (Lond) ; 4(1): 9, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216688

RESUMEN

BACKGROUND: The objective of this systematic review is to identify prognostic factors among women and their offspring affected by gestational diabetes mellitus (GDM), focusing on endpoints of cardiovascular disease (CVD) and type 2 diabetes (T2D) for women, and cardiometabolic profile for offspring. METHODS: This review included studies published in English language from January 1st, 1990, through September 30th, 2021, that focused on the above outcomes of interest with respect to sociodemographic factors, lifestyle and behavioral characteristics, traditional clinical traits, and 'omics biomarkers in the mothers and offspring during the perinatal/postpartum periods and across the lifecourse. Studies that did not report associations of prognostic factors with outcomes of interest among GDM-exposed women or children were excluded. RESULTS: Here, we identified 109 publications comprising 98 observational studies and 11 randomized-controlled trials. Findings indicate that GDM severity, maternal obesity, race/ethnicity, and unhealthy diet and physical activity levels predict T2D and CVD in women, and greater cardiometabolic risk in offspring. However, using the Diabetes Canada 2018 Clinical Practice Guidelines for studies, the level of evidence was low due to potential for confounding, reverse causation, and selection biases. CONCLUSIONS: GDM pregnancies with greater severity, as well as those accompanied by maternal obesity, unhealthy diet, and low physical activity, as well as cases that occur among women who identify as racial/ethnic minorities are associated with worse cardiometabolic prognosis in mothers and offspring. However, given the low quality of evidence, prospective studies with detailed covariate data collection and high fidelity of follow-up are warranted.


Gestational diabetes mellitus (GDM) occurs when levels of sugar in the blood are high during pregnancy. We sought to identify factors associated with short- and long-term cardiometabolic disease risk, health conditions that involve heart-related issues and complications in bodily function, among women with GDM and their offspring. We reviewed publications on factors related to type 2 diabetes (T2D) and cardiovascular disease (CVD) risk among women with GDM, and additionally assessed body composition in offspring of women with GDM. We found that GDM severity, maternal obesity, self-identified race/ethnicity, poor diet, and low physical activity levels predict postpartum T2D and CVD in the women, and unfavorable long-term cardiometabolic disease risk in offspring. The quality of evidence was poor, emphasizing a need for high-quality research capturing detailed short- and long-term outcome data to facilitate preventative interventions to improve health of women and children.

2.
Int J Obes (Lond) ; 48(2): 276-283, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38042932

RESUMEN

BACKGROUND/OBJECTIVES: Observational and experimental studies have suggested that prenatal exposure to per- and polyfluoroalkyl substances (PFAS) can increase childhood adiposity and cardiometabolic disruption. However, most previous studies have used weight-based measures that cannot distinguish between fat mass and lean mass. We evaluated associations of prenatal PFAS exposure with precisely measured body composition and cardiometabolic biomarkers in early childhood. SUBJECTS: 373 eligible mother-infant pairs in the Healthy Start longitudinal cohort. METHODS: We used multiple linear regression and Bayesian kernel machine regression models to estimate associations between five PFAS in maternal mid-pregnancy serum, and early childhood adiposity via air displacement plethysmography. Secondary outcomes included body mass index, waist circumference, and fasting serum lipids, glucose, insulin and adipokines. Models were adjusted for potential confounders and effect modification by child sex was evaluated. RESULTS: The median age of children at assessment was 4.6 years. Prenatal concentration of perfluorooctanoate (PFOA) was positively associated with percent fat mass (0.89% per log2-unit increase, 95% CI: 0.15, 1.64), while perfluorononanoate (PFNA) was positively associated with fat mass index and body mass index. Cardiometabolic markers in blood were generally not associated with prenatal PFAS in this population. Mixture models confirmed the importance of PFNA and PFOA in predicting percent fat mass, while PFNA was most important for fat mass index, body mass index, and waist circumference. There were no significant effects of the five PFAS as a mixture, potentially due to opposing effects of different PFAS. CONCLUSIONS: Our results agree with previous studies showing that prenatal serum concentrations of certain PFAS are positively associated with early childhood adiposity. Notably, associations were stronger for measures incorporating precisely measured fat mass compared to measures of body size or weight. Early life increases in adiposity may precede the development of adverse cardiometabolic health outcomes in children exposed to PFAS during gestation.


Asunto(s)
Caprilatos , Enfermedades Cardiovasculares , Contaminantes Ambientales , Fluorocarburos , Obesidad Infantil , Efectos Tardíos de la Exposición Prenatal , Niño , Embarazo , Femenino , Humanos , Preescolar , Adiposidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Teorema de Bayes , Obesidad Infantil/epidemiología , Obesidad Infantil/inducido químicamente , Enfermedades Cardiovasculares/inducido químicamente
3.
Obesity (Silver Spring) ; 32(1): 187-199, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37869908

RESUMEN

OBJECTIVE: Fetal exposures may impact offspring epigenetic signatures and adiposity. The authors hypothesized that maternal metabolic traits associate with cord blood DNA methylation, which, in turn, associates with child adiposity. METHODS: Fasting serum was obtained in 588 pregnant women (27-34 weeks' gestation), and insulin, glucose, high-density lipoprotein cholesterol, triglycerides, and free fatty acids were measured. Cord blood DNA methylation and child adiposity were measured at birth, 4-6 months, and 4-6 years. The association of maternal metabolic traits with DNA methylation (429,246 CpGs) for differentially methylated probes (DMPs) and regions (DMRs) was tested. The association of the first principal component of each DMR with child adiposity was tested, and mediation analysis was performed. RESULTS: Maternal triglycerides were associated with the most DMPs and DMRs of all traits tested (261 and 198, respectively, false discovery rate < 0.05). DMRs were near genes involved in immune function and lipid metabolism. Triglyceride-associated CpGs were associated with child adiposity at 4-6 months (32 CpGs) and 4-6 years (2 CpGs). One, near CD226, was observed at both timepoints, mediating 10% and 22% of the relationship between maternal triglycerides and child adiposity at 4-6 months and 4-6 years, respectively. CONCLUSIONS: DNA methylation may play a role in the association of maternal triglycerides and child adiposity.


Asunto(s)
Adiposidad , Metilación de ADN , Recién Nacido , Niño , Humanos , Femenino , Embarazo , Triglicéridos , Adiposidad/genética , Metabolismo de los Lípidos/genética , Sangre Fetal/metabolismo , Obesidad/metabolismo
5.
Epigenetics ; 18(1): 2254971, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37691382

RESUMEN

Background: 'Epigenetic clocks' have been developed to accurately predict chronologic gestational age and have been associated with child health outcomes in prior work.Methods: We meta-analysed results from four prospective U.S cohorts investigating the association between epigenetic age acceleration estimated using blood DNA methylation collected at birth and preschool age Childhood Behavior Checklist (CBCL) scores.Results: Epigenetic ageing was not significantly associated with CBCL total problem scores (ß = 0.33, 95% CI: -0.95, 0.28) and DSM-oriented pervasive development problem scores (ß = -0.23, 95% CI: -0.61, 0.15). No associations were observed for other DSM-oriented subscales.Conclusions: The meta-analysis results suggest that epigenetic gestational age acceleration is not associated with child emotional and behavioural functioning for preschool age group. These findings may relate to our study population, which includes two cohorts enriched for ASD and one preterm birth cohort.; future work should address the role of epigenetic age in child health in other study populations.Abbreviations: DNAm: DNA methylation; CBCL: Child Behavioral Checklist; ECHO: Environmental Influences on Child Health Outcomes; EARLI: Early Autism Risk Longitudinal Investigation; MARBLES: Markers of Autism Risk in Babies - Learning Early Signs; ELGAN: Extremely Low Gestational Age Newborns; ASD: autism spectrum disorder; BMI: body mass index; DSM: Diagnostic and Statistical Manual of Mental Disorders.


Asunto(s)
Trastorno del Espectro Autista , Nacimiento Prematuro , Preescolar , Humanos , Recién Nacido , Metilación de ADN , Epigénesis Genética , Estudios Prospectivos
6.
Obesity (Silver Spring) ; 31(8): 2090-2102, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37475691

RESUMEN

OBJECTIVE: Fat content of adipocytes derived from infant umbilical cord mesenchymal stem cells (MSCs) predicts adiposity in children through 4 to 6 years of age. This study tested the hypothesis that MSCs from infants born to mothers with obesity (Ob-MSCs) exhibit adipocyte hypertrophy and perturbations in genes regulating adipogenesis compared with MSCs from infants of mothers with normal weight (NW-MSCs). METHODS: Adipogenesis was induced in MSCs embedded in three-dimensional hydrogel structures, and cell size and number were measured by three-dimensional imaging. Proliferation and protein markers of proliferation and adipogenesis in undifferentiated and adipocyte differentiating cells were measured. RNA sequencing was performed to determine pathways linked to adipogenesis phenotype. RESULTS: In undifferentiated MSCs, greater zinc finger protein (Zfp)423 protein content was observed in Ob- versus NW-MSCs. Adipocytes from Ob-MSCs were larger but fewer than adipocytes from NW-MSCs. RNA sequencing analysis showed that Zfp423 protein correlated with mRNA expression of genes enriched for cell cycle, MSC lineage specification, inflammation, and metabolism pathways. MSC proliferation was not different before differentiation but declined faster in Ob-MSCs upon adipogenic induction. CONCLUSIONS: Ob-MSCs have an intrinsic propensity for adipocyte hypertrophy and reduced hyperplasia during adipogenesis in vitro, perhaps linked to greater Zfp423 content and changes in cell cycle pathway gene expression.


Asunto(s)
Células Madre Mesenquimatosas , Madres , Femenino , Humanos , Obesidad/genética , Obesidad/metabolismo , Diferenciación Celular/genética , Adipogénesis/genética , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción/metabolismo , Adipocitos/metabolismo , Hipertrofia/metabolismo
7.
medRxiv ; 2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37131686

RESUMEN

As part of the American Diabetes Association Precision Medicine in Diabetes Initiative (PMDI) - a partnership with the European Association for the Study of Diabetes (EASD) - this systematic review is part of a comprehensive evidence evaluation in support of the 2 nd International Consensus Report on Precision Diabetes Medicine. Here, we sought to synthesize evidence from empirical research papers published through September 1 st , 2021 to evaluate and identify prognostic conditions, risk factors, and biomarkers among women and children affected by gestational diabetes mellitus (GDM), focusing on clinical endpoints of cardiovascular disease (CVD) and type 2 diabetes (T2D) among women with a history of GDM; and adiposity and cardiometabolic profile among offspring exposed to GDM in utero. We identified a total of 107 observational studies and 12 randomized controlled trials testing the effect of pharmaceutical and/or lifestyle interventions. Broadly, current literature indicates that greater GDM severity, higher maternal body mass index, belonging to racial/ethnic minority group; and unhealthy lifestyle behaviors would predict a woman's risk of incident T2D and CVD, and an unfavorable cardiometabolic profile among offspring. However, the level of evidence is low (Level 4 according to the Diabetes Canada 2018 Clinical Practice Guidelines for diabetes prognosis) largely because most studies leveraged retrospective data from large registries that are vulnerable to residual confounding and reverse causation bias; and prospective cohort studies that may suffer selection and attrition bias. Moreover, for the offspring outcomes, we identified a relatively small body of literature on prognostic factors indicative of future adiposity and cardiometabolic risk. Future high-quality prospective cohort studies in diverse populations with granular data collection on prognostic factors, clinical and subclinical outcomes, high fidelity of follow-up, and appropriate analytical approaches to deal with structural biases are warranted.

8.
Epigenetics ; 18(1): 2179726, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36840948

RESUMEN

The placenta undergoes many changes throughout gestation to support the evolving needs of the foetus. There is also a growing appreciation that male and female foetuses develop differently in utero, with unique epigenetic changes in placental tissue. Here, we report meta-analysed sex-specific associations between gestational age and placental DNA methylation from four cohorts in the National Institutes of Health (NIH) Environmental influences on Child Health Outcomes (ECHO) Programme (355 females/419 males, gestational ages 23-42 weeks). We identified 407 cytosine-guanine dinucleotides (CpGs) in females and 794 in males where placental methylation levels were associated with gestational age. After cell-type adjustment, 55 CpGs in females and 826 in males were significant. These were enriched for biological processes critical to the immune system in females and transmembrane transport in males. Our findings are distinct between the sexes: in females, associations with gestational age are largely explained by differences in placental cellular composition, whereas in males, gestational age is directly associated with numerous alterations in methylation levels.


Asunto(s)
Metilación de ADN , Placenta , Niño , Embarazo , Humanos , Masculino , Femenino , Lactante , Placenta/metabolismo , Edad Gestacional , Epigénesis Genética , Caracteres Sexuales
9.
Obesity (Silver Spring) ; 31(1): 37-42, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36541155

RESUMEN

OBJECTIVE: In human studies, new model systems are needed for improved mechanistic investigation of developmental predisposition for metabolic disease but also to serve as benchmarks in early life prevention or intervention efforts. In this regard, human infant umbilical cord-derived mesenchymal stem cells (MSCs) are an emerging tool. However, long-term clinical relevance to in vivo markers of metabolic disease is unknown. METHODS: In a cohort of 124 mother/child dyads, this study tested the hypothesis that triglyceride content (TG) of infant MSCs undergoing adipogenesis in vitro (MSC-TG) is associated with in vivo adiposity (percent fat mass) from birth to early childhood and with fasting glucose and insulin in early childhood. RESULTS: MSC-TG was positively associated with in vivo child adiposity at birth, age 4 to 6 months, and age 4 to 6 years. MSC-TG was associated with fasting glucose, but not insulin, at 4 to 6 years. Importantly, MSC-TG explained an additional 13% variance in child adiposity at 4 to 6 years, after accounting for other established birth predictors (weight and percent fat mass at birth) and other established covariates related to child adiposity (e.g., breastfeeding exposure, physical activity). CONCLUSIONS: This work demonstrates the strength of the MSC model for predicting offspring metabolic phenotype into childhood, even when considering the important contribution of other early life risk factors.


Asunto(s)
Células Madre Mesenquimatosas , Obesidad Infantil , Recién Nacido , Niño , Femenino , Humanos , Lactante , Preescolar , Adiposidad , Glucosa/metabolismo , Obesidad Infantil/metabolismo , Ayuno , Células Madre Mesenquimatosas/metabolismo , Peso al Nacer , Índice de Masa Corporal
10.
Nutrients ; 16(1)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38201882

RESUMEN

BACKGROUND: Exposure to intrauterine obesity can disrupt clock gene rhythmicity in animal models. The aim of this pilot study was to determine if maternal obesity alters rhythmic expression of core clock in mesenchymal stem cells (MSCs) from umbilical cords of human infants born to mothers with obesity (Ob-MSC) vs. normal weight (NW-MSC). METHODS: We compared in vitro rhythmic expression patterns of core clock (BMAL1, CLOCK, PER2) and clock-output (NR1D1), components in undifferentiated Ob-MSCs (n = 3) vs. NW-MSCs (n = 3). MSCs were harvested every 2 h, following a dexamethasone shock, for 30 h. Adipogenesis or myogenesis was induced in vitro and markers of adipogenesis and fat storage were assessed, respectively. RESULTS: We detected significant rhythmicity in expression patterns of BMAL1, PER2, and NR1D1 at the group level in Ob- and NW-MSCs (p < 0.05). PER2 oscillatory amplitude was 3-fold higher in Ob-MSCs vs. NW-MSCs (p < 0.006). During adipogenesis, Ob-MSCs had higher PPARγ protein content (p = 0.04) vs. NW-MSC. During myogenesis, Ob-MSCs had higher saturated triacylglycerols (p = 0.04) vs. NW-MSC. CONCLUSION: Rhythmic expressions of BMAL1, PER2, and NR1D1 are detectable in undifferentiated MSCs. Higher PER2 oscillatory amplitude was paralleled by higher markers of fat storage during differentiation in Ob-MSCs vs. NW-MSCs, and supports that the core clock and cellular metabolism may be linked in infant MSCs.


Asunto(s)
Relojes Circadianos , Células Madre Mesenquimatosas , Embarazo , Animales , Lactante , Humanos , Femenino , Proyectos Piloto , Factores de Transcripción ARNTL/genética , Relojes Circadianos/genética , Obesidad , Expresión Génica
11.
Diabetes ; 71(8): 1649-1659, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35621990

RESUMEN

Preclinical rodent and nonhuman primate models investigating maternal obesity have highlighted the importance of the intrauterine environment in the development of insulin resistance in offspring; however, it remains unclear if these findings can be translated to humans. To investigate possible intrauterine effects in humans, we isolated mesenchymal stem cells (MSCs) from the umbilical cord tissue of infants born to mothers of normal weight or mothers with obesity. Insulin-stimulated glycogen storage was determined in MSCs undergoing myogenesis in vitro. There was no difference in insulin action based on maternal obesity. However, maternal free fatty acid (FFA) concentration, cord leptin, and intracellular triglyceride content were positively correlated with insulin action. Furthermore, MSCs from offspring born to mothers with elevated FFAs displayed elevated activation of the mTOR signaling pathway. Taken together, these data suggest that infants born to mothers with elevated lipid availability have greater insulin action in MSCs, which may indicate upregulation of growth and lipid storage pathways during periods of maternal overnutrition.


Asunto(s)
Células Madre Mesenquimatosas , Obesidad Materna , Animales , Ácidos Grasos no Esterificados/metabolismo , Femenino , Humanos , Lactante , Insulina/metabolismo , Insulina Regular Humana , Células Madre Mesenquimatosas/metabolismo , Obesidad/metabolismo , Embarazo
12.
J Clin Endocrinol Metab ; 107(8): e3353-e3365, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35511592

RESUMEN

CONTEXT: Recent preclinical data suggest exercise during pregnancy can improve the metabolic phenotype not only of the mother, but of the developing offspring as well. However, investigations in human offspring are lacking. OBJECTIVE: To characterize the effect of maternal aerobic exercise on the metabolic phenotype of the offspring's mesenchymal stem cells (MSCs). DESIGN: Randomized controlled trial. SETTING: Clinical research facility. PATIENTS: Healthy female adults between 18 and 35 years of age and ≤ 16 weeks' gestation. INTERVENTION: Mothers were randomized into 1 of 2 groups: aerobic exercise (AE, n = 10) or nonexercise control (CTRL, n = 10). The AE group completed 150 minutes of weekly moderate-intensity exercise, according to American College of Sports Medicine guidelines, during pregnancy, whereas controls attended stretching sessions. MAIN OUTCOME MEASURES: Following delivery, MSCs were isolated from the umbilical cord of the offspring and metabolic tracer and immunoblotting experiments were completed in the undifferentiated (D0) or myogenically differentiated (D21) state. RESULTS: AE-MSCs at D0 had an elevated fold-change over basal in insulin-stimulated glycogen synthesis and reduced nonoxidized glucose metabolite (NOGM) production (P ≤ 0.05). At D21, AE-MSCs had a significant elevation in glucose partitioning toward oxidation (oxidation/NOGM ratio) compared with CTRL (P ≤ 0.05). Immunoblot analysis revealed elevated complex I expression in the AE-MSCs at D21 (P ≤ 0.05). Basal and palmitate-stimulated lipid metabolism was similar between groups at D0 and D21. CONCLUSIONS: These data provide evidence of a programmed metabolic phenotype in human offspring with maternal AE during pregnancy.


Asunto(s)
Glucosa , Metabolismo de los Lípidos , Adulto , Ejercicio Físico , Femenino , Humanos , Insulina , Embarazo , Células Madre
13.
J Nutr ; 152(3): 816-825, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34850052

RESUMEN

BACKGROUND: Maternal nutritional status affects placental function, which may underlie the intrauterine origins of obesity and diabetes. The extent to which diet quality is associated with placental signaling and which specific pathways are impacted is unknown. OBJECTIVES: To examine sex-specific associations of maternal diet quality according to the Healthy Eating Index (HEI)-developed to align with recommendations from the Dietary Guidelines for Americans-with placental proteins involved in metabolism and mediators of environmental stress, inflammation, and growth factors. METHODS: Among 108 women from the Healthy Start cohort with a mean ± SD age of 29.0 ± 6.1 y and a prepregnancy BMI (in kg/m2) of 24.8 ± 5.3, we conducted multivariable linear regression analysis stratified by offspring sex. We adjusted for maternal race or ethnicity, age, education, prenatal smoking habits, and physical activity and tested for an association of maternal HEI >57 compared with ≤57 and the abundance and phosphorylation of key proteins involved in insulin/growth factor signaling; mediators of environmental stress, inflammation, and growth factors; mechanistic target of rapamycin signaling proteins; and energy sensing in placental villus samples. HEI >57 was chosen given its prior relevance among Healthy Start mother-child dyads. RESULTS: In adjusted models, HEI >57 was associated with greater abundance of insulin receptor ß (0.80; 95% CI: 0.11, 1.49) in placentas of females. In males, maternal HEI >57 was associated with greater activation and abundance of select placental nutrient-sensing proteins and environmental stress, inflammation, and growth factor proteins (S6K1Thr389/S6K1: 0.81; 95% CI: 0.21, 1.41; JNK1Thr183/Tyr185/JNK1: 0.82; 95% CI: 0.27, 1.37; JNK2Thr183/Tyr185/JNK2: 0.57; 95% CI: 0.02, 1.11). CONCLUSIONS: Higher-quality diet had sex-specific associations with placental protein abundance/phosphorylation. Given that these proteins have been correlated with neonatal anthropometry, our findings provide insight into modifiable factors and placental pathways that should be examined in future studies as potential links between maternal diet and offspring metabolic health. This trial was registered at clinicaltrials.gov as NCT02273297.


Asunto(s)
Insulina , Proteínas Gestacionales , Dieta , Femenino , Humanos , Recién Nacido , Inflamación/metabolismo , Insulina/metabolismo , Insulina Regular Humana , Masculino , Placenta/metabolismo , Embarazo , Proteínas Gestacionales/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
14.
Obesity (Silver Spring) ; 29(11): 1780-1786, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34734494

RESUMEN

OBJECTIVE: Maternal obesity increases the risks for adverse pregnancy and offspring outcomes but with large heterogeneity. This study examined changes to the maternal metabolic milieu across pregnancy in women with obesity. It identified differences between a metabolically unhealthy obesity (MUO) phenotype and a metabolically healthy obesity (MHO) phenotype, as well as the differences in offspring adiposity between the two metabolic phenotypes. METHODS: In early pregnancy, women were classified with MHO (n = 13) or MUO (n = 9) based on the presence of zero or ≥2 risk factors for metabolic syndrome, respectively (systolic blood pressure > 130 mm Hg or diastolic blood pressure > 85 mm Hg, HDL cholesterol < 50 mg/dL, LDL cholesterol ≥ 100 mg/dL, triglycerides ≥ 150 mg/dL, and glucose ≥ 100 mg/dL). Area under the pregnancy concentration curve for glucose and triglycerides measured at early (13-16 weeks), mid- (24-27 weeks), and late (35-37 weeks) pregnancy, gestational weight gain (GWG), energy expenditure, maternal fat accretion, and infant body composition were compared. RESULTS: Maternal BMI, GWG, and fat accretion did not differ between MUO and MHO. Women with MUO had a greater area under the pregnancy concentration curve for glucose (+2,170 [382] mg/dL·day, p < 0.001) and triglycerides (+12,211 [3,916] mg/dL·day, p < 0.001). There were no differences in late-pregnancy total daily energy expenditure, but activity energy expenditure was significantly lower in MUO (-403 [144] kcal). MUO offspring had greater weight (+621 [205] g, p = 0.01) and adiposity (+5.8% [2.1%], p = 0.02) at 1 week of life but showed no differences in fat-free mass. CONCLUSIONS: Independent of GWG, MUO resulted in heightened exposure of fetal fat-promoting substrates. Differing metabolic phenotypes may explain heterogeneity of offspring adiposity born to women with obesity.


Asunto(s)
Síndrome Metabólico , Obesidad Metabólica Benigna , Adiposidad , Índice de Masa Corporal , Femenino , Humanos , Obesidad , Embarazo , Factores de Riesgo
15.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34061777

RESUMEN

Exposure to maternal obesity may promote metabolic dysfunction in offspring. We used infant mesenchymal stem cells (MSCs) to experimentally examine cellular mechanisms of intergenerational health transmission. Our earlier reports show MSCs collected from infants of mothers with obesity had a dichotomous distribution in metabolic efficiency; they were either efficient (Ef-Ob) or inefficient (In-Ob) with respect to fatty acid oxidation (FAO). Here, we sought to determine if this was due to a primary defect in FAO. Accordingly, we measured FAO in myogenic differentiating MSCs under 3 conditions: (a) myogenesis alone, (b) excess fatty acid exposure, and (c) excess fatty acid exposure plus a chemical uncoupler to increase metabolic rate. Compared with normal weight and Ef-Ob MSCs, In-Ob displayed lower FAO in myogenesis alone and after fatty acid plus uncoupler, indicating In-Ob were less metabolically flexible after increasing lipid availability and metabolic rate, demonstrating a primary deficit in FAO. MSC FAO was negatively associated with fasting maternal glucose and insulin and positively associated with fasting HDL-cholesterol. MSC FAO was negatively associated with infant fat mass. These data indicate a less favorable maternal metabolic milieu, independent of maternal BMI, reduces intrinsic MSC FAO and is linked to higher infant adiposity as early as birth.


Asunto(s)
Ácidos Grasos/metabolismo , Recién Nacido/metabolismo , Células Madre Mesenquimatosas/metabolismo , Obesidad , Complicaciones del Embarazo , Efectos Tardíos de la Exposición Prenatal , Adiposidad , Peso al Nacer , Femenino , Humanos , Análisis de Flujos Metabólicos/métodos , Redes y Vías Metabólicas , Desarrollo de Músculos , Obesidad/diagnóstico , Obesidad/metabolismo , Oxidación-Reducción , Embarazo , Complicaciones del Embarazo/diagnóstico , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Efectos Tardíos de la Exposición Prenatal/metabolismo
16.
Diabetes ; 70(3): 745-751, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33414248

RESUMEN

An adverse intrauterine environment is associated with the future risk of obesity and type 2 diabetes. Changes in placental function may underpin the intrauterine origins of adult disease, but longitudinal studies linking placental function with childhood outcomes are rare. Here, we determined the abundance and phosphorylation of protein intermediates involved in insulin signaling, inflammation, cortisol metabolism, protein glycosylation, and mitochondrial biogenesis in placental villus samples from healthy mothers from the Healthy Start cohort. Using MANOVA, we tested the association between placental proteins and offspring adiposity (fat mass percentage) at birth (n = 109) and infancy (4-6 months, n = 104), and adiposity, skinfold thickness, triglycerides, and insulin in children (4-6 years, n = 66). Placental IGF-1 receptor protein was positively associated with serum triglycerides in children. GSK3ß phosphorylation at serine 9, a readout of insulin and growth factor signaling, and the ratio of phosphorylated to total JNK2 were both positively associated with midthigh skinfold thickness in children. Moreover, peroxisome proliferator-activated receptor γ coactivator (PGC)-1α abundance was positively associated with insulin in children. In conclusion, placental insulin/IGF-1 signaling, PGC-1α, and inflammation pathways were positively associated with metabolic outcomes in 4- to 6-year-old children, identifying a novel link between placental function and long-term metabolic outcomes.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Placenta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Índice de Masa Corporal , Niño , Preescolar , Femenino , Humanos , Técnicas In Vitro , Lactante , Proteína Quinasa 9 Activada por Mitógenos/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Fosforilación , Embarazo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
Environ Health Perspect ; 128(12): 127014, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33356526

RESUMEN

BACKGROUND: Per- and polyfluoroalkyl substances (PFAS) are environmentally persistent chemicals widely detected in women of reproductive age. Prenatal PFAS exposure is associated with adverse health outcomes in children. We hypothesized that DNA methylation changes may result from prenatal PFAS exposure and may be linked to offspring cardio-metabolic phenotype. OBJECTIVES: We estimated associations of prenatal PFAS with DNA methylation in umbilical cord blood. We evaluated associations of methylation at selected sites with neonatal cardio-metabolic indicators. METHODS: Among 583 mother-infant pairs in a prospective cohort, five PFAS were quantified in maternal serum (median 27 wk of gestation). Umbilical cord blood DNA methylation was evaluated using the Illumina HumanMethylation450 array. Differentially methylated positions (DMPs) were evaluated at a false discovery rate (FDR)<0.05 and differentially methylated regions (DMRs) were identified using comb-p (Sidák-adjusted p<0.05). We estimated associations between methylation at candidate DMPs and DMR sites and the following outcomes: newborn weight, adiposity, and cord blood glucose, insulin, lipids, and leptin. RESULTS: Maternal serum PFAS concentrations were below the median for females in the U.S. general population. Moderate to high pairwise correlations were observed between PFAS concentrations (ρ=0.28-0.76). Methylation at one DMP (cg18587484), annotated to the gene TJAP1, was associated with perfluorooctanoate (PFOA) at FDR< 0.05. Comb-p detected between 4 and 15 DMRs for each PFAS. Associated genes, some common across multiple PFAS, were implicated in growth (RPTOR), lipid homeostasis (PON1, PON3, CIDEB, NR1H2), inflammation and immune activity (RASL11B, RNF39), among other functions. There was suggestive evidence that two PFAS-associated loci (cg09093485, cg09637273) were associated with cord blood triglycerides and birth weight, respectively (FDR< 0.1). DISCUSSION: DNA methylation in umbilical cord blood was associated with maternal serum PFAS concentrations during pregnancy, suggesting potential associations with offspring growth, metabolism, and immune function. Future research should explore whether DNA methylation changes mediate associations between prenatal PFAS exposures and child health outcomes. https://doi.org/10.1289/EHP6888.


Asunto(s)
Exposición a Riesgos Ambientales/estadística & datos numéricos , Contaminantes Ambientales/sangre , Fluorocarburos/sangre , Biomarcadores/sangre , Metilación de ADN , Sangre Fetal/metabolismo , Humanos , Recién Nacido
18.
Int J Obes (Lond) ; 44(11): 2203-2212, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32327723

RESUMEN

OBJECTIVE: Our hypothesis was that the activity of placental nutrient-sensing pathways is associated with adiposity and metabolic health in childhood. RESEARCH DESIGN AND METHODS: Using placental villus samples from healthy mothers from the Healthy Start Study, we measured the abundance and phosphorylation of key intermediates in the mTOR, insulin, AMPK, and ER stress signaling pathways. Using multivariate multiple regression models, we tested the association between placental proteins and offspring adiposity (%fat mass) at birth (n = 109), 4-6 months (n = 104), and 4-6 years old (n = 64), adjusted for offspring sex and age. RESULTS: Placental mTORC1 phosphorylation was positively associated with adiposity at birth (R2 = 0.13, P = 0.009) and 4-6 years (R2 = 0.15, P = 0.046). The mTORC2 target PKCα was positively associated with systolic blood pressure at 4-6 years (ß = 2.90, P = 0.005). AMPK phosphorylation was positively associated with adiposity at birth (ß = 2.32, P = 0.023), but the ratio of phosphorylated to total AMPK was negatively associated with skinfold thickness (ß = -2.37, P = 0.022) and body weight (ß = -2.92, P = 0.005) at 4-6 years. CONCLUSIONS: This is the first report of associations between key placental protein activity measures and longitudinal child outcomes at various life stages. Our data indicate that AMPK and mTOR signaling are linked to cardiometabolic measures at birth and 4-6 years, providing novel insight into potential mechanisms underpinning how metabolic signaling in the placenta is associated with future risk of cardiovascular disease.


Asunto(s)
Adiposidad , Placenta/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP/metabolismo , Peso al Nacer , Niño , Preescolar , Colorado , Estrés del Retículo Endoplásmico , Femenino , Humanos , Lactante , Recién Nacido , Insulina/metabolismo , Estudios Longitudinales , Masculino , Nutrientes , Fosforilación , Embarazo , Grosor de los Pliegues Cutáneos , Serina-Treonina Quinasas TOR/metabolismo
19.
Nutrients ; 12(3)2020 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-32183350

RESUMEN

Overweight and obesity accompanies up to 70% of pregnancies and is a strong risk factor for offspring metabolic disease. Maternal obesity-associated inflammation and lipid profile are hypothesized as important contributors to excess offspring liver and skeletal muscle lipid deposition and oxidative stress. Here, we tested whether dams expressing the fat-1 transgene, which endogenously converts omega-6 (n-6) to omega-3 (n-3) polyunsaturated fatty acid, could protect wild-type (WT) offspring against high-fat diet induced weight gain, oxidative stress, and disrupted mitochondrial fatty acid oxidation. Despite similar body mass at weaning, offspring from fat-1 high-fat-fed dams gained less weight compared with offspring from WT high-fat-fed dams. In particular, WT males from fat-1 high-fat-fed dams were protected from post-weaning high-fat diet induced weight gain, reduced fatty acid oxidation, or excess oxidative stress compared with offspring of WT high-fat-fed dams. Adult offspring of WT high-fat-fed dams exhibited greater skeletal muscle triglycerides and reduced skeletal muscle antioxidant defense and redox balance compared with offspring of WT dams on control diet. Fat-1 offspring were protected from the reduced fatty acid oxidation and excess oxidative stress observed in offspring of WT high-fat-fed dams. These results indicate that a maternal fat-1 transgene has protective effects against offspring liver and skeletal muscle lipotoxicity resulting from a maternal high-fat diet, particularly in males. Altering maternal fatty acid composition, without changing maternal dietary composition or weight gain with high-fat feeding, may highlight important strategies for n-3-based prevention of developmental programming of obesity and its complications.


Asunto(s)
Proteínas de Caenorhabditis elegans , Dieta Alta en Grasa/efectos adversos , Ácido Graso Desaturasas , Exposición Materna , Obesidad , Estrés Oxidativo/genética , Transgenes , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Ácido Graso Desaturasas/biosíntesis , Ácido Graso Desaturasas/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Obesidad/prevención & control , Embarazo
20.
Methods Mol Biol ; 1978: 355-368, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31119674

RESUMEN

Spectrophotometric analysis of metabolic enzyme activity from homogenized tissues is a valuable method for investigating mitochondrial content and capacity. Enzyme activity is normally measured in single cuvette spectrophotometers, requiring a large sample volume and low throughput. Here, we describe microplate assays for high-throughput analysis of mitochondrial enzymes citrate synthase, ß-hydroxyacyl CoA dehydrogenase, aconitase, and mitochondrial electron transport system (ETS) complexes I, II, III, and IV.


Asunto(s)
Complejo I de Transporte de Electrón/química , Enzimas/química , Mitocondrias/enzimología , Espectrofotometría/métodos , Aconitato Hidratasa/química , Transporte de Electrón/genética , Enzimas/metabolismo , Mitocondrias/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...