Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Matern Child Health J ; 21(6): 1367-1376, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28138825

RESUMEN

Objective Obesity in adults is associated with inflammation and oxidative stress. Whether or not this phenotype is reflected in human milk (HM) composition, or may impact infant growth remains unknown. We investigated whether HM from overweight/obese (OW/Ob) mothers exhibited higher concentrations of inflammatory cytokines and markers of oxidative stress. We also correlated these bioactive components with infant growth patterns. Methods This was an observational cohort of 56 breastfeeding mothers and their infants [33 normal weight (NW) and 23 OW/Ob]. Infants were followed until 6 months of age and HM collected at 2-weeks and 4-months. Results Markers of oxidative stress, 8-hydroxy-deoxyguanosine (8OHdG) and 4-hydroxynonenol (HNE), decreased in HM over time (p < 0.001) and did not differ between NW and OW/Ob women. Concentrations of inflammatory cytokines, IL-6, IL-8, and TNF-α, were all inter-correlated (p < 0.001) but did not differ between NW and OW/Ob women. HM fat, protein, lactose, and total calories did not differ between NW and OW/Ob women. Infant growth patterns did not differ by group. In a model of infant weight-for-length-Z score trajectory, there was a significant interaction between both lactose and 8OHdG with maternal group: HM lactose and 8OHdG concentrations were both positively associated with increases in WLZ trajectory only among infants breastfed by OW/Ob mothers. Conclusions for Practice HM composition was relatively stable between NW and OW/Ob women. In exclusively breastfed infants, HM concentrations of lactose and 8OHdG, a marker of oxidative stress, may contribute to regulation of infant weight gain, especially among infants of OW/Ob women.


Asunto(s)
Biomarcadores/análisis , Índice de Masa Corporal , Leche Humana/química , Madres , Lactancia Materna , Desarrollo Infantil , Preescolar , Estudios de Cohortes , Citocinas/análisis , Femenino , Humanos , Lactante , Lactancia/fisiología , Estudios Longitudinales , Obesidad/metabolismo , Obesidad/fisiopatología , Sobrepeso/fisiopatología , Estrés Oxidativo
2.
J Nutr ; 145(3): 514-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25733467

RESUMEN

BACKGROUND: The zinc content of maize, a major global food staple, is generally insufficient alone to meet the requirements of young children. OBJECTIVES: The primary objective of this study was to determine whether substitution of biofortified maize (34 µg zinc/g grain) for control maize (21 µg zinc/g) was adequate to meet zinc physiologic requirements in young children for whom maize was the major food staple. A secondary objective was to compare total daily zinc absorption when maize flour was fortified with zinc oxide to a total concentration of 60 µg zinc/g. METHODS: Participants included 60 rural Zambian children with a mean age of 29 mo who were randomly assigned to receive 1 of 3 maize types (control, biofortified, or fortified) all of which were readily consumed (>100 g on 1 d). Total daily zinc intake (from maize and low-zinc relish) was determined from duplicate diet collections. Multiplication by fractional absorption of zinc, measured by a dual isotope ratio technique, determined the total daily zinc absorption on the day the test meals were given. RESULTS: The mean ± SD total daily zinc intake (milligrams per day) from the biofortified maize (5.0 ± 2.2) was higher (P < 0.0001) than for the control maize (2.3 ± 0.9). Intake of zinc from the fortified maize (6.3 ± 2.6) did not differ from the biofortified maize. Fractional absorption of zinc from control maize (0.28 ± 0.10) did not differ from the biofortified maize (0.22 ± 0.06). Total daily absorption of zinc (milligrams per day) from the biofortified maize (1.1 ± 0.5) was higher (P = 0.0001) than for the control maize (0.6 ± 0.2), but did not differ from the fortified maize (1.2 ± 0.4). CONCLUSIONS: These results indicate that feeding biofortified maize can meet zinc requirements and provide an effective dietary alternative to regular maize for this vulnerable population. This trial was registered at clinicaltrials.gov as NCT02208635.


Asunto(s)
Dieta , Alimentos Fortificados , Población Rural , Zea mays/química , Zinc/administración & dosificación , Zinc/farmacocinética , Preescolar , Estudios Transversales , Femenino , Harina/análisis , Humanos , Lactante , Masculino
3.
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
4.
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
5.
Child Obes ; 13(2): 102-110, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27936906

RESUMEN

BACKGROUND: Adolescents who are obese are at high risk of developing obstructive sleep apnea (OSA). Although there is clear evidence associating OSA with metabolic dysfunction in adults, the evidence is less clear cut with adolescents. The purpose of this review was determine the association of sleep apnea with dyslipidemia, insulin resistance, cardiovascular disease risk, nonalcoholic fatty liver disease, and difficulty with weight loss in adolescents. METHODS: A systematic literature review using PubMed, Scopus, CINAHL, Google Scholar, and PsycINFO was performed and articles were screened and reviewed with an a priori protocol. RESULTS: Sixteen articles were included in qualitative synthesis and 10 were included in meta-analysis. Results from the meta-analysis indicate that OSA in adolescents is associated with greater risk of dyslipidemia, insulin resistance, and hypertension. CONCLUSIONS: Although obesity leads to increased metabolic risk, OSA appears to independently increase metabolic impairment. Adolescents with obesity should be frequently screened for OSA to determine need for treatment and reduce this metabolic burden.


Asunto(s)
Síndrome Metabólico/metabolismo , Obesidad Infantil/etiología , Obesidad Infantil/metabolismo , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/metabolismo , Adolescente , Glucemia/metabolismo , Índice de Masa Corporal , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Humanos , Resistencia a la Insulina , Síndrome Metabólico/epidemiología , Síndrome Metabólico/etiología , Obesidad Infantil/epidemiología , Factores de Riesgo , Apnea Obstructiva del Sueño/epidemiología , Estados Unidos/epidemiología
6.
Sci Rep ; 7(1): 18095, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29273781

RESUMEN

The intrauterine period is a critical time wherein developmental exposure can influence risk for chronic disease including childhood obesity. Using umbilical cord-derived mesenchymal stem cells (uMSC) from offspring born to normal-weight and obese mothers, we tested the hypothesis that changes in infant body composition over the first 5 months of life correspond with differences in cellular metabolism and transcriptomic profiles at birth. Higher long-chain acylcarnitine concentrations, lipid transport gene expression, and indicators of oxidative stress in uMSC-adipocytes were related to higher adiposity at 5 months of age. In uMSC-myocytes, lower amino acid concentrations and global differential gene expression for myocyte growth, amino acid biosynthesis, and oxidative stress were related to lower infant percent fat-free mass at 5 months of age, particularly in offspring of obese mothers. This is the first evidence of human infant adipocyte- or myocyte-related alterations in cellular metabolic pathways that correspond with increased adiposity and lower fat-free mass in early infancy. These pathways might reflect the effects of an adverse maternal metabolic environment on the fetal metabolome and genome. Our findings suggest that programmed differences in infant stem cell metabolism correspond with differences in body composition in early life, a known contributor to obesity risk.


Asunto(s)
Adiposidad/fisiología , Peso al Nacer/fisiología , Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Índice de Masa Corporal , Diferenciación Celular , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Metabolómica , Estrés Oxidativo/fisiología
7.
Mol Metab ; 6(11): 1503-1516, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29107296

RESUMEN

OBJECTIVE: Infants born to mothers with obesity have greater adiposity, ectopic fat storage, and are at increased risk for childhood obesity and metabolic disease compared with infants of normal weight mothers, though the cellular mechanisms mediating these effects are unclear. METHODS: We tested the hypothesis that human, umbilical cord-derived mesenchymal stem cells (MSCs) from infants born to obese (Ob-MSC) versus normal weight (NW-MSC) mothers demonstrate altered fatty acid metabolism consistent with adult obesity. In infant MSCs undergoing myogenesis in vitro, we measured cellular lipid metabolism and AMPK activity, AMPK activation in response to cellular nutrient stress, and MSC DNA methylation and mRNA content of genes related to oxidative metabolism. RESULTS: We found that Ob-MSCs exhibit greater lipid accumulation, lower fatty acid oxidation (FAO), and dysregulation of AMPK activity when undergoing myogenesis in vitro. Further experiments revealed a clear phenotype distinction within the Ob-MSC group where more severe MSC metabolic perturbation corresponded to greater neonatal adiposity and umbilical cord blood insulin levels. Targeted analysis of DNA methylation array revealed Ob-MSC hypermethylation in genes regulating FAO (PRKAG2, ACC2, CPT1A, SDHC) and corresponding lower mRNA content of these genes. Moreover, MSC methylation was positively correlated with infant adiposity. CONCLUSIONS: These data suggest that greater infant adiposity is associated with suppressed AMPK activity and reduced lipid oxidation in MSCs from infants born to mothers with obesity and may be an important, early marker of underlying obesity risk.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metilación de ADN , Ácidos Grasos/metabolismo , Obesidad/metabolismo , Obesidad Infantil/epidemiología , Obesidad Infantil/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Acetil-CoA Carboxilasa/genética , Adulto , Carnitina O-Palmitoiltransferasa/genética , Ácidos Grasos/genética , Femenino , Humanos , Lactante , Recién Nacido , Metabolismo de los Lípidos , Masculino , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/metabolismo , Madres , Desarrollo de Músculos/fisiología , Obesidad/enzimología , Obesidad/genética , Oxidación-Reducción , Obesidad Infantil/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal , Cordón Umbilical/citología , Cordón Umbilical/metabolismo , Adulto Joven
8.
Diabetes ; 65(3): 647-59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26631736

RESUMEN

Maternal obesity increases the risk for pediatric obesity; however, the molecular mechanisms in human infants remain poorly understood. We hypothesized that mesenchymal stem cells (MSCs) from infants born to obese mothers would demonstrate greater potential for adipogenesis and less potential for myogenesis, driven by differences in ß-catenin, a regulator of MSC commitment. MSCs were cultured from the umbilical cords of infants born to normal-weight (prepregnancy [pp] BMI 21.1 ± 0.3 kg/m(2); n = 15; NW-MSCs) and obese mothers (ppBMI 34.6 ± 1.0 kg/m(2); n = 14; Ob-MSCs). Upon differentiation, Ob-MSCs exhibit evidence of greater adipogenesis (+30% Oil Red O stain [ORO], +50% peroxisome proliferator-activated receptor (PPAR)-γ protein; P < 0.05) compared with NW-MSCs. In undifferentiated cells, total ß-catenin protein content was 10% lower and phosphorylated Thr41Ser45/total ß-catenin was 25% higher (P < 0.05) in Ob-MSCs versus NW-MSCs (P < 0.05). Coupled with 25% lower inhibitory phosphorylation of GSK-3ß in Ob-MSCs (P < 0.05), these data suggest greater ß-catenin degradation in Ob-MSCs. Lithium chloride inhibition of GSK-3ß increased nuclear ß-catenin content and normalized nuclear PPAR-γ in Ob-MSCs. Last, ORO in adipogenic differentiating cells was positively correlated with the percent fat mass in infants (r = 0.475; P < 0.05). These results suggest that altered GSK-3ß/ß-catenin signaling in MSCs of infants exposed to maternal obesity may have important consequences for MSC lineage commitment, fetal fat accrual, and offspring obesity risk.


Asunto(s)
Adipogénesis/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Células Madre Mesenquimatosas/metabolismo , Obesidad/metabolismo , PPAR gamma/metabolismo , Complicaciones del Embarazo/metabolismo , beta Catenina/metabolismo , Adulto , Diferenciación Celular , Células Cultivadas , Estudios de Cohortes , Femenino , Glucógeno Sintasa Quinasa 3 beta , Humanos , Estudios Longitudinales , Masculino , Células Madre Mesenquimatosas/fisiología , Desarrollo de Músculos/fisiología , Obesidad Infantil , Embarazo , Cordón Umbilical/citología
9.
PLoS One ; 11(7): e0159575, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27414406

RESUMEN

The cellular mechanisms whereby excess maternal nutrition during pregnancy increases adiposity of the offspring are not well understood. However, nicotinamide (NAM), a fundamental micronutrient that is important in energy metabolism, has been shown to regulate adipogenesis through inhibition of SIRT1. Here we tested three novel hypotheses: 1) NAM increases the adipogenic response of human umbilical cord tissue-derived mesenchymal stem cells (MSCs) through a SIRT1 and PPARγ pathway; 2) lipid potentiates the NAM-enhanced adipogenic response; and 3) the adipogenic response to NAM is associated with increased percent fat mass (%FM) among neonates. MSCs were derived from the umbilical cord of 46 neonates born to non-obese mothers enrolled in the Healthy Start study. Neonatal %FM was measured using air displacement plethysmography (Pea Pod) shortly after birth. Adipogenic differentiation was induced for 21 days in the 46 MSC sets under four conditions, +NAM (3mM)/-lipid (200 µM oleate/palmitate mix), +NAM/+lipid, -NAM/+lipid, and vehicle-control (-NAM/-lipid). Cells incubated in the presence of NAM had significantly higher PPARγ protein (+24%, p <0.01), FABP4 protein (+57%, p <0.01), and intracellular lipid content (+51%, p <0.01). Lipid did not significantly increase either PPARγ protein (p = 0.98) or FABP4 protein content (p = 0.82). There was no evidence of an interaction between NAM and lipid on adipogenic response of PPARγ or FABP4 protein (p = 0.99 and p = 0.09). In a subset of 9 MSC, SIRT1 activity was measured in the +NAM/-lipid and vehicle control conditions. SIRT1 enzymatic activity was significantly lower (-70%, p <0.05) in the +NAM/-lipid condition than in vehicle-control. In a linear model with neonatal %FM as the outcome, the percent increase in PPARγ protein in the +NAM/-lipid condition compared to vehicle-control was a significant predictor (ß = 0.04, 95% CI 0.01-0.06, p <0.001). These are the first data to support that chronic NAM exposure potentiates adipogenesis in human MSCs in-vitro, and that this process involves PPARγ and SIRT1.


Asunto(s)
Adipogénesis/efectos de los fármacos , Adiposidad/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Niacinamida/farmacología , Cordón Umbilical/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Recién Nacido/metabolismo , PPAR gamma/metabolismo , Pletismografía , Reacción en Cadena de la Polimerasa , Sirtuina 1/metabolismo , Cordón Umbilical/citología
10.
Am J Clin Nutr ; 103(5): 1291-300, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27140533

RESUMEN

BACKGROUND: Increased maternal body mass index (BMI) is a robust risk factor for later pediatric obesity. Accumulating evidence suggests that human milk (HM) may attenuate the transfer of obesity from mother to offspring, potentially through its effects on early development of the infant microbiome. OBJECTIVES: Our objective was to identify early differences in intestinal microbiota in a cohort of breastfeeding infants born to obese compared with normal-weight (NW) mothers. We also investigated relations between HM hormones (leptin and insulin) and both the taxonomic and functional potentials of the infant microbiome. DESIGN: Clinical data and infant stool and fasting HM samples were collected from 18 NW [prepregnancy BMI (in kg/m(2)) <24.0] and 12 obese (prepregnancy BMI >30.0) mothers and their exclusively breastfed infants at 2 wk postpartum. Infant body composition at 2 wk was determined by air-displacement plethysmography. Infant gastrointestinal microbes were estimated by using 16S amplicon and whole-genome sequencing. HM insulin and leptin were determined by ELISA; short-chain fatty acids (SCFAs) were measured in stool samples by using gas chromatography. Power was set at 80%. RESULTS: Infants born to obese mothers were exposed to 2-fold higher HM insulin and leptin concentrations (P < 0.01) and showed a significant reduction in the early pioneering bacteria Gammaproteobacteria (P = 0.03) and exhibited a trend for elevated total SCFA content (P < 0.06). Independent of maternal prepregnancy BMI, HM insulin was positively associated with both microbial taxonomic diversity (P = 0.03) and Gammaproteobacteria (e.g., Enterobacteriaceae; P = 0.04) and was negatively associated with Lactobacillales (e.g., Streptococcaceae; P = 0.05). Metagenomic analysis showed that HM leptin and insulin were associated with decreased bacterial proteases, which are implicated in intestinal permeability, and reduced concentrations of pyruvate kinase, a biomarker of pediatric gastrointestinal inflammation. CONCLUSION: Our results indicate that, although maternal obesity may adversely affect the early infant intestinal microbiome, HM insulin and leptin are independently associated with beneficial microbial metabolic pathways predicted to increase intestinal barrier function and reduce intestinal inflammation. This trial was registered at clinicaltrials.gov as NCT01693406.


Asunto(s)
Microbioma Gastrointestinal , Insulina/análisis , Leptina/análisis , Leche Humana/química , Adulto , Biomarcadores/sangre , Composición Corporal , Índice de Masa Corporal , Lactancia Materna , Estudios de Cohortes , Estudios Transversales , Ácidos Grasos Volátiles/análisis , Heces/química , Femenino , Gammaproteobacteria/aislamiento & purificación , Humanos , Lactante , Lactobacillales/aislamiento & purificación , Modelos Lineales , Masculino , Análisis Multivariante , Obesidad/sangre , Obesidad/prevención & control , Pletismografía , Piruvato Quinasa/sangre , Factores de Riesgo
11.
Physiol Behav ; 104(1): 98-104, 2011 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-21539852

RESUMEN

Endogenous opioids are involved in the hedonic aspects of eating. Opioid impairments and alterations have been implicated in the pathophysiology of bulimia nervosa and binge eating disorder. Specific contributions by Bartley G. Hoebel have furthered the understanding how cyclical caloric restriction and intermittent optional access to sugar solutions result in opioid-like forebrain neural alterations and dependency in rodents. The present study sought to investigate caudal brainstem and nodose ganglion mu-opioid receptor mRNA alterations in a rodent model of dietary-induced binge eating of sweetened fat (vegetable shortening blended with 10% sucrose). Five groups (n=7 or 8) of adult female Sprague Dawley rats were exposed to various dietary conditions for 6 weeks. As measured by in situ hybridization, there was reduced (approximately 25% from naive) mu-opioid receptor mRNA in the nucleus of the solitary tract (NTS) in the binge access group, which had intermittent calorie restriction and optional limited access to the sweetened fat. A similar reduction in expression was demonstrated in the continuous access group, which has unlimited optional sweetened fat and an obese phenotype. In the nodose ganglion, mu-opioid receptor mRNA was increased (approximately 30% from groups with sweetened fat access) in rats with intermittent caloric restriction alone. Our findings and the body of work from the Hoebel laboratory suggest that dietary-induced binge eating can consequentially alter opioidergic forebrain and hindbrain feeding-related neural pathways. Future work is needed to determine whether similar alterations are involved in the maintenance and progression of binge eating and other related eating pathologies.


Asunto(s)
Tronco Encefálico/metabolismo , Bulimia/metabolismo , Vías Nerviosas/metabolismo , Ganglio Nudoso/metabolismo , Receptores Opioides mu/metabolismo , Animales , Bulimia/genética , Bulimia/fisiopatología , Dieta , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Femenino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA