scholarly journals Erratum to: Milky Ways: Effects of Maternal Obesity on Human Milk Composition and Childhood Obesity Risk. Am J Clin Nutr 2021;113(4):772–74.

2021 ◽  
Vol 113 (6) ◽  
pp. 1715-1715
2019 ◽  
Vol 110 (1) ◽  
pp. 111-120 ◽  
Author(s):  
Elvira Isganaitis ◽  
Sarah Venditti ◽  
Tucker J Matthews ◽  
Carles Lerin ◽  
Ellen W Demerath ◽  
...  

ABSTRACTBackgroundMaternal obesity is a risk factor for childhood obesity; this is a major public health concern given that ∼40% of pregnant women are either overweight or obese. Whether differences in milk composition in lean compared with obese women contribute to childhood obesity is unclear.ObjectivesWe aimed to analyze relationships between maternal obesity and human milk metabolites, infant body composition, and postnatal weight gain.MethodsThis was a prospective study in which mothers intending to breastfeed exclusively, and their newborn infants, were enrolled at delivery (n = 35 mother–infant pairs). We excluded mothers with diabetes, other medical conditions, or pregnancy complications. Participants were grouped by maternal prepregnancy BMI <25 (lean) or ≥25 kg/m2 (overweight/obese). We analyzed infant body composition by dual-energy X-ray absorptiometry and used untargeted liquid chromatography–gas chromatography–mass spectrometry to measure the milk content of 275 metabolites at 1 and 6 mo postpartum.ResultsAt 1 mo postpartum, 10 metabolites differed between overweight/obese and lean groups with nominal P < 0.05, but none was altered with a false discovery rate <0.25. Many differentially abundant metabolites belonged to the same chemical class; e.g., 4/10 metabolites were nucleotide derivatives, and 3/10 were human milk oligosaccharides. Milk adenine correlated positively with both continuously distributed maternal BMI and with infant adiposity and fat accrual. Analysis of milk composition at 6 mo postpartum revealed 20 differentially abundant metabolites (P < 0.05) in overweight/obese compared with lean women, including 6 metabolites with a false discovery rate of <0.25. At both 1 and 6 mo, human milk abundance of 1,5-anhydroglucitol, which has not previously been described in milk, was positively associated with maternal BMI.ConclusionsMaternal obesity is associated with changes in the human milk metabolome. While only a subset of metabolites correlated with both maternal and infant weight, these point to potential milk-dependent mechanisms for mother–child transmission of obesity. This trial was registered at www.clinicaltrials.gov as NCT02535637.


2020 ◽  
pp. 000992282097100
Author(s):  
James Gannon ◽  
Allison J. Pollock ◽  
David B. Allen ◽  
Pamela J. Kling

Children obese at the age of 5 years are at greater risk of lifelong obesity. Because certain risks of obesity can be identified in early infancy, a tool for obesity risk prediction in early life would be clinically useful. We investigated predictors of obesity risk in a novel, prospectively collected healthy birth cohort recruited for demographic risks to develop iron deficiency at 1 year, a cohort leveraged because risk factors for iron deficiency and obesity overlap. Obesity at the age of 5 years was defined as age- and sex-specific body mass index Z-score ( zBMI) >2SD. For each child, obesity risk factors were summed. Of 10 total risk factors, the following 4 key risks were identified: maternal obesity, maternal diabetes, large for gestational age, or breastfeeding <6 months. Childhood obesity was predicted by either ≥3 total number of risks ( P < .033), any key risk ( P < .002), or summing key risks ( P < .0001). In clinical practice, summing early life risk factors may be a useful strategy for preemptive counseling.


2021 ◽  
Vol 5 (Supplement_2) ◽  
pp. 818-818
Author(s):  
Clark Sims ◽  
Audrey , Martinez ◽  
Aline Andres

Abstract Objectives Human milk (HM) is impacted by both maternal obesity and diet. HM from women with obesity has higher fat, leptin and insulin concentrations. Additionally, maternal dietary fat intake is associated with increased HM fat content. The objective of this study was to determine the impact of a Mediterranean meal plan on the composition of HM from women with obesity. Methods In this pilot study, thirteen women with obesity (body mass index (BMI) ≥ 30 kg/m2) enrolled at five months post-partum and followed a provided Mediterranean meal plan focused on reducing fat intake and increasing unsaturated fat and fiber intake for four weeks. Participants’ diet was assessed using Nutrition Data System for Research. HM was collected at baseline and after the 4-week intervention and the concentrations of metabolites, hormones and cytokines were assayed. Wilcoxon signed-rank tests and analysis of variance were used to assess changes in participant diet and HM composition. Summary statistics are presented as mean ± standard deviation. Results The participants’ Healthy Eating Index Score, a measure of diet quality, significantly improved (38.3 ± 5.13 vs. 78.2 ± 6.75, p &lt; 0.001) and total fat intake was significantly lower (99.6 ± 23.3 g vs. 66.9 ± 12.0 g/day, p &lt; 0.001) after the intervention. Human milk leptin (694 ± 464 pg/ml vs. 437 ± 324 pg/ml, p = 0.007) and tumor necrosis factor α (0.515 ± 0.267 pg/ml vs. 0.310 ± 0.127 pg/ml, p = 0.010) concentrations were lower post intervention, whereas HM macronutrient composition was unchanged. The concentrations of several individual human milk oligosaccharides (HMO) and total HMOs (7720 ± 797 nmol/ml vs. 6986 ± 940 nmol/ml, p = 0.049), as well as HM tyrosine concentration (18.2 ± 6.16 nmol/ml vs. 14.5 ± 4.69 nmol/ml, p = 0.005) were significantly lower post intervention. Conclusions The composition of HM from women with obesity can be modulated by short-term adherence to a Mediterranean meal pattern. These findings merit further studies that use longer interventions and examine the impact of any changes in HM composition on infant growth and development. Modifying HM composition via a dietary intervention may provide a novel strategy to promote child development and health. Funding Sources USDA ARS #6026-51,000-010-05S and #6026-51,000-012-06S, NIH/NIDDK R01DK107516, Arkansas Children's Research Institute/Arkansas Biosciences Institute GR037121.


2020 ◽  
Vol 112 (5) ◽  
pp. 1228-1239
Author(s):  
Jessica L Saben ◽  
Clark R Sims ◽  
Brian D Piccolo ◽  
Aline Andres

ABSTRACT Background Human milk composition is altered by maternal obesity. The association between milk metabolites and infant outcomes has not been thoroughly investigated. Objectives This study aimed to quantify maternal adiposity-related differences in the human milk metabolome and to identify metabolites associated with infant adiposity during the first 6 mo postpartum using untargeted metabolomics. Method Maternal anthropometrics were assessed ≤14 weeks of gestation. Human milk samples were collected at 0.5 mo (n = 159), 2 mo (n = 131), and 6 mo (n = 94) postpartum from normal weight (NW, BMI = 18.5–24.9 kg/m2) and obese (OB, BMI &gt;30 kg/m2) mothers. GC-time-of-flight-MS was used to identify metabolic signatures that discriminate NW and OB women. Partial least squared (PLS)-discriminant analysis, and PLS-regression models were assessed to examine relations between metabolites and maternal BMI and fat mass. Metabolites altered by maternal obesity were used in linear mixed effect models to predict infant adiposity. Results Multivariate modeling identified 23, 17, and 10 metabolites that described maternal adiposity indices at 0.5 mo, 2 mo, and 6 mo postpartum, respectively. Monosaccharides and sugar alcohols were the most representative annotated metabolite classes that were increased in milk from OB women and included: mannose, ribose, lyxose, lyxitol (0.5 mo); mannose, ribitol, glycerol, isothreonic acid, lyxitol (2 mo); lyxitol and isothreonic acid (6 mo). Other discriminant metabolites included: 1-monostearin, xylonolactone, shikimic acid, pseudo uridine, and dodecanol (0.5 mo); N-acetyl-D-hexosamine and fumaric acid (2 mo); uric acid and tyrosine (6 mo). Mannose, lyxitol, and shikimic acid predicted higher infant adiposity over the first 6 mo of life. Conclusions This study reports on 1 of the largest cohorts to date examining the metabolic profiles in human milk comparing NW and OB women. Maternal adiposity was associated with increased amounts of milk nonglucose monosaccharides. Human milk metabolomics may be useful in predicting infant adiposity. These trials were registered at www.clinicaltrials.gov as NCT01131117 and NCT02125149.


Nutrients ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 299
Author(s):  
Junilla K. Larsen ◽  
Lars Bode

Animal studies have consistently demonstrated that maternal obesity and a high-fat diet during lactation enhances obesity risk in the offspring. However, less is known about these potential obesogenic programming effects in obese humans. We propose three important pathways that may explain obesogenic programming effects of human breastmilk. First, human milk components and hormones may directly affect child eating and satiety characteristics. Second, human milk constituents can affect child microbiota that, in turn, may influence child eating and weight outcomes. Third, human milk composition may affect child eating and weight outcomes through flavor exposure. We reviewed a few very recent findings from well-powered longitudinal or experimental human research with regard to these three pathways. Moreover, we provide a research agenda for future intervention research with the overarching aim to prevent excessive pediatric weight gain during lactation and beyond. The ideas presented in this paper may represent important “black box” constructs that explain obesogenic programming effects during lactation. It should be noted, however, that given the scarcity of studies, findings should be seen as working hypotheses to further test in future research.


Author(s):  
Syaza Y. Binte Abu Bakar ◽  
Malinda Salim ◽  
Andrew J. Clulow ◽  
Kevin Nicholas ◽  
Ben J. Boyd
Keyword(s):  

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Sara Jalali-Farahani ◽  
Parisa Amiri ◽  
Bita Lashkari ◽  
Leila Cheraghi ◽  
Farhad Hosseinpanah ◽  
...  

Abstract Background Parental weight is studied as an important determinant of childhood obesity; however, obesity-related metabolic abnormalities have been less considered as determinants of childhood obesity. This study aimed to investigate the association between maternal obesity phenotypes and incidence of obesity in their offspring. Methods This longitudinal study was conducted within the framework of the Tehran Lipid and Glucose Study. A total of 2151 non-obese children who had complete parental information were followed for incidence of obesity over a mean of 148.7 ± 34.7 months. Obesity in children was defined using the World Health Organization criteria. Maternal body mass index (BMI) was classified into three categories: normal weight, overweight and obese. Dysmetabolic status was considered as having metabolic syndrome or diabetes. Metabolic syndrome and diabetes were defined according to the Joint Interim Statement and American diabetes association criteria, respectively. Considering maternal BMI categories and metabolic status, six obesity phenotypes were defined as followed: 1) normal weight and normal metabolic status, 2) overweight and normal metabolic status, 3) obese and normal metabolic status, 4) normal weight and dysmetabolic status, 5) overweight and dysmetabolic status, and 6) obese and dysmetabolic status. The association between maternal obesity phenotypes and incidence of obesity in children was studied using Cox proportional regression hazard model. Results In male offspring, the risk of incidence of obesity significantly increased in those with maternal obesity phenotypes including overweight/normal metabolic: 1.75(95% CI: 1.10–2.79), obese/normal metabolic: 2.60(95%CI: 1.51–4.48), overweight/dysmetabolic: 2.34(95%CI: 1.35–4.03) and obese/dysmetabolic: 3.21(95%CI: 1.94–5.03) compared to the normal weight/normal metabolic phenotype. Similarly, in girls, the risk of incidence of obesity significantly increased in offspring with maternal obesity phenotypes including overweight/normal metabolic: 2.39(95%CI: 1.46–3.90), obese/normal metabolic: 3.55(95%CI: 1.94–6.46), overweight/dysmetabolic: 1.92(95%CI: 1.04–3.52) and obese/dysmetabolic: 3.89(95%CI: 2.28–6.64) compared to normal weight/normal metabolic phenotype. However, maternal normal weight/dysmetabolic phenotype did not significantly change the risk of obesity in both male and female offspring. Conclusion Except for normal weight/dysmetabolic phenotype, all maternal obesity phenotypes had significant prognostic values for incidence of offspring obesity with the highest risk for obese/dysmetabolic phenotype. This study provides valuable findings for identifying the first line target groups for planning interventions to prevent childhood obesity.


Life ◽  
2021 ◽  
Vol 11 (2) ◽  
pp. 114
Author(s):  
Diana Escuder-Vieco ◽  
Juan M. Rodríguez ◽  
Irene Espinosa-Martos ◽  
Nieves Corzo ◽  
Antonia Montilla ◽  
...  

Holder pasteurization (HoP; 62.5 °C, 30 min) is commonly used to ensure the microbiological safety of donor human milk (DHM) but diminishes its nutritional properties. A high-temperature short-time (HTST) system was designed as an alternative for human milk banks. The objective of this study was to evaluate the effect of this HTST system on different nutrients and the bile salt stimulated lipase (BSSL) activity of DHM. DHM was processed in the HTST system and by standard HoP. Macronutrients were measured with a mid-infrared analyzer. Lactose, glucose, myo-inositol, vitamins and lipids were assayed using chromatographic techniques. BSSL activity was determined using a kit. The duration of HTST treatment had a greater influence on the nutrient composition of DHM than did the tested temperature. The lactose concentration and the percentage of phospholipids and PUFAs were higher in HTST-treated than in raw DHM, while the fat concentration and the percentage of monoacylglycerides and SFAs were lower. Other nutrients did not change after HTST processing. The retained BSSL activity was higher after short HTST treatment than that following HoP. Overall, HTST treatment resulted in better preservation of the nutritional quality of DHM than HoP because relevant thermosensitive components (phospholipids, PUFAs, and BSSL) were less affected.


Author(s):  
Parul Christian ◽  
Emily R Smith ◽  
Sun Eun Lee ◽  
Ashley J Vargas ◽  
Andrew A Bremer ◽  
...  

ABSTRACT Critical advancement is needed in the study of human milk as a biological system that intersects and interacts with myriad internal (maternal biology) and external (diet, environment, infections) factors and its plethora of influences on the developing infant. Human-milk composition and its resulting biological function is more than the sum of its parts. Our failure to fully understand this biology in a large part contributes to why the duration of exclusive breastfeeding remains an unsettled science (if not policy). Our current understanding of human-milk composition and its individual components and their functions fails to fully recognize the importance of the chronobiology and systems biology of human milk in the context of milk synthesis, optimal timing and duration of feeding, and period of lactation. The overly simplistic, but common, approach to analyzing single, mostly nutritive components of human milk is insufficient to understand the contribution of either individual components or the matrix within which they exist to both maternal and child health. There is a need for a shift in the conceptual approach to studying human milk to improve strategies and interventions to support better lactation, breastfeeding, and the full range of infant feeding practices, particularly for women and infants living in undernourished and infectious environments. Recent technological advances have led to a rising movement towards advancing the science of human-milk biology. Herein, we describe the rationale and critical need for unveiling the multifunctionality of the various nutritional, nonnutritional, immune, and biological signaling pathways of the components in human milk that drive system development and maturation, growth, and development in the very early postnatal period of life. We provide a vision and conceptual framework for a research strategy and agenda to change the field of human-milk biology with implications for global policy, innovation, and interventions.


Sign in / Sign up

Export Citation Format

Share Document