Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.508
Filter
1.
Genes (Basel) ; 15(6)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38927748

ABSTRACT

Infant consumption of human milk (HM) is associated with a reduced risk of overweight and obesity, but the reasons for this relationship are not completely understood. There is emerging evidence that micro RNAs (miRNAs) regulate infant development and metabolism, but the associations between HM miRNAs and infant growth remain poorly understood. We examined the relationship between HM miRNA consumption and infant obesity in 163 mother-infant dyads to determine (1) if miRNA profiles differentiate infants with obesity, and (2) if individual miRNAs accurately predicted infant obesity status at one year of age. Infant obesity was categorized as weight-for-length (WFL) Z scores or conditional weight gain (CWG) in the 95th percentile. HM miRNA profile was associated with infant age (r2 = 6.4%, p = 0.001), but not maternal obesity status (r2 = 1.5%, p = 0.87) or infant weight status (WFL Z-score) at birth (r2 = 0.6%, p = 0.4), 1 month (r2 = 0.5%, p = 0.6), or 4 months (r2 = 0.8%, p = 0.2). Nine HM miRNAs were associated with either 12-month CWG or 12-month WFL Z scores. Among these 9 miRNAs, miR-224-5p remained significant in a logistic regression model that accounted for additional demographic factors (estimate = -27.57, p = 0.004). These findings suggest involvement of HM miRNAs and particularly miR-224-5p in infant growth, warranting further investigation. To our knowledge, this is the largest study of HM miRNAs and early-life obesity and contributes to the understanding of the relationship between HM miRNAs and infant growth.


Subject(s)
MicroRNAs , Milk, Human , Humans , Milk, Human/metabolism , Milk, Human/chemistry , Female , MicroRNAs/genetics , Infant , Male , Adult , Infant, Newborn , Obesity/genetics , Pediatric Obesity/genetics , Breast Feeding
2.
Sci Rep ; 14(1): 14685, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38918476

ABSTRACT

Human milk oligosaccharides (HMOs) promote adequate intestinal microbiota development and favor the immune system's maturation and cognitive development. In addition to non-modifiable factors, HMOs composition can be influenced by other factors like body mass index and eating habits, but the reports are discrepant. The aim of this work was to describe the correlation between maternal factors and HMOs concentration in colostrum in 70 women from northeastern Mexico categorized into women with normal weight and women with overweight or obesity. The absolute concentration of six HMOs were significantly lower in women with overweight or obesity compared to women with normal weight (LNFPI p = 0.0021, 2'-FL p = 0.0304, LNT p = 0.0492, LNnT p = 0.00026, 3'-SL p = 0.0476, 6'-SL p = 0.00041). Another main finding was that the frequency of consumption of food groups such as vegetables, fruits and meats was positively correlated to specific HMOs (Poblano chili and 2'-FL; rs = 0.702, p = 0.0012; Orange or tangerine and 3-FL; rs = 0.428, p = 0.0022; Chicken and 2'-FL; rs = 0.615, p = 0.0039). This study contributes to the elucidation of how maternal factors influence the composition of HMOs and opens possibilities for future research aimed at mitigating overweight or obesity, consequently improving the quality of human milk.


Subject(s)
Breast Feeding , Feeding Behavior , Milk, Human , Oligosaccharides , Humans , Milk, Human/chemistry , Milk, Human/metabolism , Female , Mexico , Oligosaccharides/analysis , Adult , Obesity/metabolism , Body Mass Index , Colostrum/chemistry , Colostrum/metabolism , Overweight , Young Adult
3.
PLoS One ; 19(6): e0305421, 2024.
Article in English | MEDLINE | ID: mdl-38870243

ABSTRACT

Human milk is optimal for infant nutrition. However, many mothers cease breastfeeding because of low milk supply (LMS). It is difficult to identify mothers at risk for LMS because its biologic underpinnings are not fully understood. Previously, we demonstrated that milk micro-ribonucleic acids (miRNAs) may be related to LMS. Transforming growth factor beta (TGFß) also plays an important role in mammary involution and may contribute to LMS. We performed a longitudinal cohort study of 139 breastfeeding mothers to test the hypothesis that milk levels of TGFß would identify mothers with LMS. We explored whether TGFß impacts the expression of LMS-related miRNAs in cultured human mammary epithelial cells (HMECs). LMS was defined by maternal report of inadequate milk production, and confirmed by age of formula introduction and infant weight trajectory. Levels of TGF-ß1 and TGF-ß2 were measured one month after delivery. There was a significant relationship between levels of TGF-ß1 and LMS (X2 = 8.92, p = 0.003) on logistic regression analysis, while controlling for lactation stage (X2 = 1.28, p = 0.25), maternal pre-pregnancy body mass index (X2 = 0.038, p = 0.84), and previous breastfeeding experience (X2 = 7.43, p = 0.006). The model accounted for 16.8% of variance in the data (p = 0.005) and correctly predicted LMS for 84.6% of mothers (22/26; AUC = 0.72). Interactions between TGF-ß1 and miR-22-3p displayed significant effect on LMS status (Z = 2.67, p = 0.008). Further, incubation of HMECs with TGF-ß1 significantly reduced mammary cell number (t = -4.23, p = 0.003) and increased levels of miR-22-3p (t = 3.861, p = 0.008). Interactions between TGF-ß1 and miR-22-3p may impact mammary function and milk levels of TGF-ß1 could have clinical utility for identifying mothers with LMS. Such information could be used to provide early, targeted lactation support.


Subject(s)
Breast Feeding , MicroRNAs , Milk, Human , Transforming Growth Factor beta1 , Humans , Female , Milk, Human/metabolism , Transforming Growth Factor beta1/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Adult , Lactation , Transforming Growth Factor beta2/metabolism , Longitudinal Studies , Epithelial Cells/metabolism , Infant , Mothers , Infant, Newborn , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology
4.
Nat Food ; 5(6): 480-490, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872016

ABSTRACT

Human milk oligosaccharides (HMOs) are a diverse class of carbohydrates which support the health and development of infants. The vast health benefits of HMOs have made them a commercial target for microbial production; however, producing the approximately 200 structurally diverse HMOs at scale has proved difficult. Here we produce a diversity of HMOs by leveraging the robust carbohydrate anabolism of plants. This diversity includes high-value and complex HMOs, such as lacto-N-fucopentaose I. HMOs produced in transgenic plants provided strong bifidogenic properties, indicating their ability to serve as a prebiotic supplement with potential applications in adult and infant health. Technoeconomic analyses demonstrate that producing HMOs in plants provides a path to the large-scale production of specific HMOs at lower prices than microbial production platforms. Our work demonstrates the promise in leveraging plants for the low-cost and sustainable production of HMOs.


Subject(s)
Milk, Human , Oligosaccharides , Plants, Genetically Modified , Oligosaccharides/metabolism , Humans , Milk, Human/metabolism , Milk, Human/chemistry , Plants, Genetically Modified/genetics , Prebiotics , Photosynthesis
5.
EBioMedicine ; 104: 105182, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38838470

ABSTRACT

BACKGROUND: Human milk oligosaccharides (HMOs), their determinants, infant gut microbiota and health are under extensive research; however, seldom jointly addressed. Leveraging data from the HELMi birth cohort, we investigated them collectively, considering maternal and infant secretor status. METHODS: HMO composition in breastmilk collected 3 months postpartum (n = 350 mothers) was profiled using high-performance liquid chromatography. Infant gut microbiota taxonomic and functional development was studied at 3, 6, and 12 months (n = 823 stool samples) via shotgun metagenomic sequencing, focusing on HMO metabolism via glycoside hydrolase (GH) analysis. Maternal and infant secretor statuses were identified through phenotyping and genotyping, respectively. Child health, emphasizing allergies and antibiotics as proxies for infectious diseases, was recorded until 2 years. FINDINGS: Mother's parity, irritable bowel syndrome, gestational diabetes, and season of milk collection associated with HMO composition. Neither maternal nor infant secretor status associated with infant gut microbiota, except for a few taxa linked to individual HMOs. Analysis stratified for birth mode revealed distinct patterns between the infant gut microbiota and HMOs. Child health parameters were not associated to infant or maternal secretor status. INTERPRETATION: This comprehensive exploration unveils intricate links between secretor genotype, maternal factors, HMO composition, infant microbiota, and child health. Understanding these nuanced relationships is paramount for refining strategies to optimize early life nutrition and its enduring impact on long-term health. FUNDING: Sweet Crosstalk EU H2020 MSCA ITN, Academy of Finland, Mary and Georg C. Ehrnrooth Foundation, Päivikki and Sakari Sohlberg Foundation, and Tekes.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Oligosaccharides , Parity , Seasons , Humans , Milk, Human/chemistry , Milk, Human/metabolism , Oligosaccharides/metabolism , Oligosaccharides/analysis , Female , Finland , Infant , Birth Cohort , Metagenomics/methods , Pregnancy , Infant, Newborn , Adult , Metagenome , Male , Feces/microbiology
6.
Front Endocrinol (Lausanne) ; 15: 1415630, 2024.
Article in English | MEDLINE | ID: mdl-38938519

ABSTRACT

Objective: Endocannabinoids and their N-acyl-ethanolamines (NAEs) and 2monoacyl-glycerols (2-MAGs) congeners are involved in the central and peripheral regulation of energy homeostasis, they are present in human milk and are associated with obesity. Infants exposed in utero to gestational diabetes mellitus (GDM) are more likely to develop obesity. The objective of this cross-sectional study is to compare the profile of eCBome mediators in milk of women with gestational diabetes (GDM+) and without (GDM-) and to assess the association with offspring growth. The hypothesis is that the eCBome of GDM+ human milk is altered and associated with a difference in infant growth. Methods: Circulating eCBome mediators were measured by LC-MS/MS in human milk obtained at 2 months postpartum from GDM+ (n=24) and GDM- (n=29) women. Infant weight and height at 2 months were obtained from the child health record. Z-scores were calculated. Results: Circulating Npalmitoylethanolamine (PEA) was higher in human milk of GDM+ women than in GDM- women (4.9 ± 3.2 vs. 3.3 ± 1.7, p=0.04). Higher levels were also found for several 2monoacyl-glycerols (2-MAGs) (p<0.05). The levels of NAEs (ß=-4.6, p=0.04) and especially non-omega-3 NAEs (B=-5.6, p=0.004) in human milk were negatively correlated with weight-for-age z-score of GDM+ offspring. Conclusion: The profile of eCBome mediators in human milk at 2 months postpartum was different in GDM+ compared to GDM- women and was associated with GDM+ offspring growth at 2 months. Clinical trial registration: ClinicalTrials.gov, identifier (NCT04263675 and NCT02872402).


Subject(s)
Diabetes, Gestational , Endocannabinoids , Milk, Human , Adult , Female , Humans , Infant , Infant, Newborn , Male , Pregnancy , Child Development/physiology , Cross-Sectional Studies , Diabetes, Gestational/metabolism , Diabetes, Gestational/blood , Endocannabinoids/blood , Endocannabinoids/metabolism , Milk, Human/chemistry , Milk, Human/metabolism
7.
Front Public Health ; 12: 1389513, 2024.
Article in English | MEDLINE | ID: mdl-38841677

ABSTRACT

Background: Peripartum cardiomyopathy (PPCM) is a common cause of heart failure (HF) in the peripartum. Some medications are considered safe while breastfeeding. However, sacubitril/valsartan (Entresto), while efficacious, is not recommended in breastfeeding women due to concerns about adverse infant development, and no published data suggest otherwise. Objectives: This study aimed to assess the transfer of sacubitril/valsartan into human milk and evaluate the infant's risk of drug exposure. Methods: The InfantRisk Human Milk Biorepository released samples and corresponding health information from five breastfeeding maternal-infant dyads exposed to sacubitril/valsartan. Sacubitril, valsartan, and LBQ657 (sacubitril active metabolite) concentrations were determined using liquid chromatography-mass spectrometry (LC/MS/MS) from timed samples 0, 1, 2, 4, 6, 8, 10, and 12 h following medication administration at steady state conditions. Results: Valsartan levels were below the detection limit of 0.19 ng/mL in all milk samples. Sacubitril was measurable in all milk samples of the five participants, peaking 1 h after drug administration at a mean concentration of 1.52 ng/mL for a total infant dose of 0.00049 mg/kg/12 h and a relative infant dose (RID) calculated at 0.01%. The maximum concentration of its active metabolite LBQ657 in the milk samples was observed 4 h after medication administration and declined over the remaining 12-h dosing interval, for an average concentration of 9.5 ng/mL. The total infant dose was 0.00071 mg/kg/12 h, and the RID was 0.22%. Two mothers reported continuing to breastfeed while taking sacubitril/valsartan; both mothers stated observing no negative effects in their breastfed infants. Conclusion: The transfer of sacubitril/valsartan into human milk is minimal. These concentrations are unlikely to pose a significant risk to breastfeeding infants, with a combined calculated RID of <0.25%, which is far lower than the industry safety standards (RID <10%).


Subject(s)
Aminobutyrates , Biphenyl Compounds , Breast Feeding , Drug Combinations , Milk, Human , Valsartan , Humans , Milk, Human/chemistry , Milk, Human/metabolism , Female , Aminobutyrates/analysis , Adult , Chromatography, Liquid , Pregnancy , Tandem Mass Spectrometry , Infant, Newborn , Tetrazoles , Infant , Angiotensin Receptor Antagonists/administration & dosage , Cardiomyopathies
8.
Clin Nutr ESPEN ; 62: 144-156, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38901936

ABSTRACT

BACKGROUND & AIMS: Human milk (HM) is a complete food that meets the nutritional and energy demands of the newborns. It contains numerous bioactive components, including functional proteins. Variations in HM energy and lipid content have already been reported related to the newborn's sex, but differences between protein profiles are still scarce. This work aimed to identify differences between HM proteins produced by mothers of female and male newborns, in the lactation stages of colostrum and mature milk, and the metabolic pathways involved. METHODS: A total of 98 HM samples were collected from 39 lactating women and classified according to the newborn's sex, stages of lactation, and three mothers' age groups, and evaluated about protein concentration and one-dimensional electrophoretic profile. Next, to assess samples with the greatest differences, the HM proteins regarding the newborn's sex and the stages of lactation were compared using nano-LC-MS/MS, in 24 HM samples randomly rearranged into four groups: female and male infants, and colostrum and mature milk. Functional classification, metabolic pathways, and protein interaction networks were analyzed by Gene Ontology, KEGG, and STRING, respectively. RESULTS: The soluble protein content of HM decreased throughout lactation, with differences regarding isolated factors, such as mothers' age group, child's sex and stages of lactation, and also in terms of their interactions. A total of 146 proteins were identified, 42 of which showed different abundances over the sexes of newborns and 53 between the stages of lactation. In general, proteins related to metabolic processes were up-regulated for mothers of male infants and in the mature stage of lactation, while proteins related to defense were up-regulated in mothers of female infants and in the colostrum phase. CONCLUSION: This study indicated that there are differentiated and specific nutritional and defense needs of newborns, by sex and by lactation phase, which is highly relevant for a more appropriate supply of food to infants receiving HM from donor mothers.


Subject(s)
Colostrum , Lactation , Milk Proteins , Milk, Human , Humans , Female , Milk, Human/chemistry , Milk, Human/metabolism , Lactation/physiology , Male , Infant, Newborn , Milk Proteins/analysis , Adult , Colostrum/chemistry , Sex Factors , Breast Feeding , Young Adult , Tandem Mass Spectrometry
9.
Sci Rep ; 14(1): 13448, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38862662

ABSTRACT

Human milk (HM) components affect immune cell toll-like receptor 4 (TLR4) signaling. However, studies examining the immunomodulatory impacts of HM on TLR4 signaling in intestinal epithelial cells (IECs) are limited. This study utilized both a TLR4 reporter cell line and a Caco-2 IEC model to examine the effects of HM on lipopolysaccharide (LPS)-induced TLR4 activation and cytokine responses, respectively. Additionally, we performed fast protein liquid chromatography and mass spectrometry to identify a HM component that contributes to the effect of HM on LPS/TLR4 signaling. HM enhances LPS-induced TLR4 signaling as well as LPS-induced IEC gene expression of pro-inflammatory cytokines and negative regulators of NF-κB. Human serum albumin (HSA) present in HM contributes to these effects. HSA within HM synergizes with LPS to induce IEC gene expression of pro-inflammatory cytokines and negative regulators of NF-κB. Altogether, this study provides mechanistic evidence behind the immunomodulatory function of HM on IECs, which may contribute to an enhanced immune response in breast-fed neonates.


Subject(s)
Cytokines , Lipopolysaccharides , Milk, Human , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Humans , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Milk, Human/metabolism , Milk, Human/chemistry , Lipopolysaccharides/pharmacology , Cytokines/metabolism , Caco-2 Cells , Signal Transduction/drug effects , NF-kappa B/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Gene Expression Regulation/drug effects
10.
Sci Total Environ ; 941: 173511, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38825210

ABSTRACT

4-Hydroxychlorothalonil (4-OH CHT), the main metabolite of chlorothalonil and the most widely used fungicide, has been frequently detected in human samples during monitoring. 4-OH CHT may exhibit higher toxicity and persistence in the environment compared to its prototype. In this study, a total of 540 paired serum and breast milk samples from pregnant women in three provinces in China were monitored for contaminant residues. 4-OH CHT was analyzed in the samples using ultra high-performance liquid chromatography - high-resolution mass spectrometry with a detection limit of 20 ng/L. The study investigated the effects of demographic factors, such as BMI, region of residence, and education level, on the levels of 4-OH CHT residues in serum and breast milk. Among the three provinces, the highest median concentration of 4-OH CHT in serum samples was observed in Hebei (1.04 × 103 ng/L), while the highest median concentration of 4-OH CHT in breast milk samples was observed in Hubei and Guangdong (491 ng/L). Multiple linear regression was used to investigate the significant positive correlation between 4-OH CHT in serum and breast milk (p = 0.000) after adjusting for personal characteristics. Based on this, the study further explored the influencing factors of transfer efficiencies (TEs) in conjunction with the individual TEs and the personal characteristics of the participants. Our results demonstrated that the age of the volunteers and their exercise habits had an effect on TEs, but further studies are needed to determine whether exercise leads to an increase in TEs.


Subject(s)
Fungicides, Industrial , Milk, Human , Nitriles , Milk, Human/chemistry , Milk, Human/metabolism , Humans , Female , China , Nitriles/analysis , Adult , Cross-Sectional Studies , Fungicides, Industrial/analysis , Pregnancy , Maternal Exposure/statistics & numerical data , Cities , Environmental Monitoring , Environmental Pollutants/metabolism , Environmental Pollutants/analysis
11.
J Clin Psychopharmacol ; 44(4): 337-344, 2024.
Article in English | MEDLINE | ID: mdl-38739007

ABSTRACT

PURPOSE/BACKGROUND: Zuranolone is a positive allosteric modulator of both synaptic and extrasynaptic γ-aminobutyric acid type A receptors and a neuroactive steroid approved as an oral, once-daily, 14-day treatment course for adults with postpartum depression in the United States. This study assessed zuranolone transfer into breast milk. METHODS/PROCEDURES: Healthy, nonpregnant, lactating adult female participants received once-daily 30 mg zuranolone from day (D)1 through D5 in this phase 1 open-label study. The relative infant dose (RID; weight-adjusted proportion of the maternal dose in breast milk over 24 hours) for 30 mg zuranolone was assessed at D5. An RID for 50 mg zuranolone was estimated using a simulation approach across a range of infant ages and weights. FINDINGS/RESULTS: Of 15 enrolled participants (mean age, 30.1 years), 14 completed the study. The mean RID for 30 mg zuranolone at D5 was 0.357%; the mean steady-state milk volume over D3 to D5 decreased from baseline by 8.3%. Overall unbound zuranolone in plasma was low (≤0.49%). Plasma concentrations peaked at D5 before decreasing in a biexponential manner. There was strong concordance between the temporal profiles of zuranolone concentrations in plasma and breast milk. The estimated mean RID for 50 mg zuranolone based on a milk intake of 200 mL/kg per day was 0.984%. All treatment-emergent adverse events reported by participants were mild, the most common being dizziness (n = 3). IMPLICATIONS/CONCLUSIONS: Zuranolone transfer into the breast milk of healthy, nonpregnant, lactating adult female participants was low; the estimated RID for 50 mg zuranolone was <1%, well below the <10% threshold generally considered compatible with breastfeeding.


Subject(s)
Lactation , Milk, Human , Humans , Female , Adult , Milk, Human/metabolism , Lactation/drug effects , Lactation/metabolism , Young Adult , Healthy Volunteers , Pregnanolone , Pyrazoles
12.
Gut Microbes ; 16(1): 2359729, 2024.
Article in English | MEDLINE | ID: mdl-38816999

ABSTRACT

Early life environment influences mammalian brain development, a growing area of research within the Developmental Origins of Health and Disease framework, necessitating a deeper understanding of early life factors on children's brain development. This study introduces a mouse model, LAO1 knockout mice, to investigate the relationship between breast milk, the gut microbiome, and brain development. The results reveal that breast milk's reactive oxygen species (ROS) are vital in shaping the neonatal gut microbiota. Decreased hydrogen peroxide (H2O2) levels in milk disrupt the gut microbiome and lead to abnormal metabolite production, including D-glucaric acid. This metabolite inhibits hippocampal myelin formation during infancy, potentially contributing to behavioral abnormalities observed in adulthood. These findings suggest that H2O2 in breast milk is crucial for normal gut microbiota formation and brain development, with implications for understanding and potentially treating neurodevelopmental disorders in humans.


Subject(s)
Animals, Newborn , Gastrointestinal Microbiome , Hydrogen Peroxide , Mice, Knockout , Milk, Human , Myelin Sheath , Animals , Female , Male , Mice , Animals, Newborn/growth & development , Brain/metabolism , Brain/growth & development , Hippocampus/metabolism , Hippocampus/growth & development , Hydrogen Peroxide/metabolism , Mice, Inbred C57BL , Milk, Human/chemistry , Milk, Human/metabolism , Myelin Sheath/metabolism , Reactive Oxygen Species/metabolism
13.
J Agric Food Chem ; 72(19): 10665-10678, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38691667

ABSTRACT

This review explores the role of microorganisms and metabolites in human breast milk and their impact on neonatal health. Breast milk serves as both a primary source of nutrition for newborns and contributes to the development and maturation of the digestive, immunological, and neurological systems. It has the potential to reduce the risks of infections, allergies, and asthma. As our understanding of the properties of human milk advances, there is growing interest in incorporating its benefits into personalized infant nutrition strategies, particularly in situations in which breastfeeding is not an option. Future infant formula products are expected to emulate the composition and advantages of human milk, aligning with an evolving understanding of infant nutrition. The long-term health implications of human milk are still under investigation.


Subject(s)
Infant Health , Microbiota , Milk, Human , Milk, Human/chemistry , Milk, Human/metabolism , Humans , Infant , Infant, Newborn , Female , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Infant Nutritional Physiological Phenomena , Breast Feeding
14.
Food Res Int ; 186: 114317, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729709

ABSTRACT

Lipids play a pivotal role in the nutrition of preterm infants, acting as a primary energy source. Due to their underdeveloped gastrointestinal systems, lipid malabsorption is common, leading to insufficient energy intake and slowed growth. Therefore, it is critical to explore the reasons behind the low lipid absorption rate in formulas for preterm infants. This study utilized a simulated in intro gastrointestinal digestion model to assess the differences in lipid digestion between preterm human milk and various infant formulas. Results showed that the fatty acid release rates for formulas IF3, IF5, and IF7 were 58.90 %, 56.58 %, and 66.71 %, respectively, lower than human milk's 72.31 %. The primary free fatty acids (FFA) and 2-monoacylglycerol (2-MAG) released during digestion were C14:0, C16:0, C18:0, C18:1n-9, and C18:2n-6, in both human milk and formulas. Notably, the higher release of C16:0 in formulas may disrupt fatty acid balance, impacting lipid absorption. Further investigations are necessary to elucidate lipid absorption differences, which will inform the optimization of lipid content in preterm infant formulas.


Subject(s)
Digestion , Infant Formula , Infant, Premature , Milk, Human , Milk, Human/chemistry , Milk, Human/metabolism , Humans , Infant Formula/chemistry , Infant, Newborn , Fatty Acids/analysis , Fatty Acids/metabolism , Lipids/analysis , Fatty Acids, Nonesterified/analysis , Fatty Acids, Nonesterified/metabolism , Lipid Metabolism , Gastrointestinal Tract/metabolism , Models, Biological , Monoglycerides/metabolism , Monoglycerides/analysis , Dietary Fats/metabolism , Dietary Fats/analysis
15.
Clin Pharmacokinet ; 63(5): 561-588, 2024 May.
Article in English | MEDLINE | ID: mdl-38748090

ABSTRACT

Human milk is a remarkable biofluid that provides essential nutrients and immune protection to newborns. Breastfeeding women consuming medications could pass the drug through their milk to neonates. Drugs can be transferred to human milk by passive diffusion or active transport. The physicochemical properties of the drug largely impact the extent of drug transfer into human milk. A comprehensive understanding of the physiology of human milk formation, composition of milk, mechanisms of drug transfer, and factors influencing drug transfer into human milk is critical for appropriate selection and use of medications in lactating women. Quantification of drugs in the milk is essential for assessing the safety of pharmacotherapy during lactation. This can be achieved by developing specific, sensitive, and reproducible analytical methods using techniques such as liquid chromatography coupled with mass spectrometry. The present review briefly discusses the physiology of human milk formation, composition of human milk, mechanisms of drug transfer into human milk, and factors influencing transfer of drugs from blood to milk. We further expand upon and critically evaluate the existing analytical approaches/assays used for the quantification of drugs in human milk.


Subject(s)
Milk, Human , Humans , Milk, Human/chemistry , Milk, Human/metabolism , Pharmaceutical Preparations/metabolism , Female , Lactation/metabolism , Breast Feeding , Infant, Newborn , Chromatography, Liquid/methods , Mass Spectrometry/methods
16.
Nat Commun ; 15(1): 3851, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719803

ABSTRACT

Current guidelines advise against primaquine treatment for breastfeeding mothers to avoid the potential for haemolysis in infants with G6PD deficiency. To predict the haemolytic risk, the amount of drug received from the breast milk and the resulting infant drug exposure need to be characterised. Here, we develop a pharmacokinetic model to describe the drug concentrations in breastfeeding women using venous, capillary, and breast milk data. A mother-to-infant model is developed to mimic the infant feeding pattern and used to predict their drug exposures. Primaquine and carboxyprimaquine exposures in infants are <1% of the exposure in mothers. Therefore, even in infants with the most severe G6PD deficiency variants, it is highly unlikely that standard doses of primaquine (0.25-1 mg base/kg once daily given to the mother for 1-14 days) would cause significant haemolysis. After the neonatal period, primaquine should not be restricted for breastfeeding women (Clinical Trials Registration: NCT01780753).


Subject(s)
Antimalarials , Breast Feeding , Lactation , Milk, Human , Primaquine , Humans , Female , Primaquine/pharmacokinetics , Primaquine/administration & dosage , Antimalarials/pharmacokinetics , Antimalarials/administration & dosage , Infant , Milk, Human/chemistry , Milk, Human/metabolism , Adult , Infant, Newborn , Hemolysis/drug effects , Models, Biological
17.
PLoS One ; 19(5): e0292997, 2024.
Article in English | MEDLINE | ID: mdl-38728264

ABSTRACT

BACKGROUND: Current research suggests that energy transfer through human milk influences infant nutritional development and initiates metabolic programming, influencing eating patterns into adulthood. To date, this research has predominantly been conducted among women in high income settings and/or among undernourished women. We will investigate the relationship between maternal body composition, metabolic hormones in human milk, and infant satiety to explore mechanisms of developmental satiety programming and implications for early infant growth and body composition in Samoans; a population at high risk and prevalence for overweight and obesity. Our aims are (1) to examine how maternal body composition influences metabolic hormone transfer from mother to infant through human milk, and (2) to examine the influences of maternal metabolic hormone transfer and infant feeding patterns on early infant growth and satiety. METHODS: We will examine temporal changes in hormone transfers to infants through human milk in a prospective longitudinal cohort of n = 80 Samoan mother-infant dyads. Data will be collected at three time points (1, 3, & 4 months postpartum). At each study visit we will collect human milk and fingerpick blood samples from breastfeeding mother-infant dyads to measure the hormones leptin, ghrelin, and adiponectin. Additionally, we will obtain body composition measurements from the dyad, observe breastfeeding behavior, conduct semi-structured interviews, and use questionnaires to document infant hunger and feeding cues and satiety responsiveness. Descriptive statistics, univariate and multivariate analyses will be conducted to address each aim. DISCUSSION: This research is designed to advance our understanding of variation in the developmental programming of satiety and implications for early infant growth and body composition. The use of a prospective longitudinal cohort alongside data collection that utilizes a mixed methods approach will allow us to capture a more accurate representation on both biological and cultural variables at play in a population at high risk of overweight and obesity.


Subject(s)
Body Composition , Milk, Human , Humans , Milk, Human/metabolism , Milk, Human/chemistry , Female , Infant , Prospective Studies , Longitudinal Studies , Leptin/blood , Leptin/metabolism , Adiponectin/blood , Adiponectin/metabolism , Adult , Ghrelin/blood , Ghrelin/metabolism , Child Development/physiology , Male , Breast Feeding , Infant Nutritional Physiological Phenomena , Satiation/physiology , Mothers
18.
J Nutr ; 154(7): 2006-2013, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38718924

ABSTRACT

BACKGROUND: In lactating women, iodine metabolism is regulated and maintained by the kidneys and mammary glands. Limited research exists on how iodine absorbed by lactating women is distributed between the kidneys and breasts. OBJECTIVES: This study aimed to accurately evaluate the total iodine intake (TII), urinary iodine excretion (UIE), and breast milk iodine excretion (BMIE) in lactating women and explore the relationship between TII and total iodine excretion (TIE). METHODS: A 7-d iodine metabolism study was conducted on 41 lactating women with a mean age of 30 y in Yuncheng and Gaoqing, China, from December 2021 to August 2023. TII and TIE were calculated by measuring the iodine content in food, water, 24-h urine, feces, and breast milk. The urinary iodine excretion rate (UIER), breast milk iodine excretion rate (BMIER), and partitioning of iodine excretion between urine and breast milk were determined. RESULTS: Iodine metabolism studies were performed for 285 d. The median TII and TIE values were 255 and 263 µg/d, respectively. With an increase in TII, UIER, and BMIER, the UIE and BMIE to TII ratio exhibited a downward trend. The median UIER, BMIER, and proportion of iodine excreted in urine and breast milk were 51.5%, 38.5%, 52%, and 37%, respectively. When the TII was <120 µg/d, the BMIER decreased with the increase of the TII (ß: -0.90; 95% confidence interval: -1.08, -0.72). CONCLUSIONS: When maternal iodine intake is low, the proportion in breast milk increases, ensuring sufficient iodine nutrition for infants. In addition, the UIE of lactating women with adequate iodine concentrations is higher than their BMIE. This study was registered at clinicaltrials.gov as NCT04492657.


Subject(s)
Iodine , Lactation , Milk, Human , Humans , Iodine/urine , Iodine/metabolism , Milk, Human/chemistry , Milk, Human/metabolism , Female , Lactation/metabolism , Adult , China
19.
Nutrients ; 16(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38794769

ABSTRACT

Several metabolites of the essential amino acid tryptophan have emerged as key players in gut homeostasis through different cellular pathways, particularly through metabolites which can activate the aryl hydrocarbon receptor (AHR). This study aimed to map the metabolism of tryptophan in early life and investigate the effects of specific metabolites on epithelial cells and barrier integrity. Twenty-one tryptophan metabolites were measured in the feces of full-term and preterm neonates as well as in human milk and formula. The ability of specific AHR metabolites to regulate cytokine-induced IL8 expression and maintain barrier integrity was assessed in Caco2 cells and human fetal organoids (HFOs). Overall, higher concentrations of tryptophan metabolites were measured in the feces of full-term neonates compared to those of preterm ones. Within AHR metabolites, indole-3-lactic acid (ILA) was significantly higher in the feces of full-term neonates. Human milk contained different levels of several tryptophan metabolites compared to formula. Particularly, within the AHR metabolites, indole-3-sulfate (I3S) and indole-3-acetic acid (IAA) were significantly higher compared to formula. Fecal-derived ILA and milk-derived IAA were capable of reducing TNFα-induced IL8 expression in Caco2 cells and HFOs in an AHR-dependent manner. Furthermore, fecal-derived ILA and milk-derived IAA significantly reduced TNFα-induced barrier disruption in HFOs.


Subject(s)
Feces , Milk, Human , Receptors, Aryl Hydrocarbon , Tryptophan , Humans , Receptors, Aryl Hydrocarbon/metabolism , Milk, Human/metabolism , Milk, Human/chemistry , Caco-2 Cells , Tryptophan/metabolism , Infant, Newborn , Feces/chemistry , Indoleacetic Acids/metabolism , Female , Infant, Premature , Interleukin-8/metabolism , Indoles/pharmacology , Infant Formula , Organoids/metabolism , Basic Helix-Loop-Helix Transcription Factors
20.
Nutrients ; 16(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38794660

ABSTRACT

Breastfeeding is the most appropriate source of a newborn's nutrition; among the plethora of its benefits, its modulation of circadian rhythmicity with melatonin as a potential neuroendocrine transducer has gained increasing interest. Transplacental transfer assures melatonin provision for the fetus, who is devoid of melatonin secretion. Even after birth, the neonatal pineal gland is not able to produce melatonin rhythmically for several months (with an even more prolonged deficiency following preterm birth). In this context, human breast milk constitutes the main natural source of melatonin: diurnal dynamic changes, an acrophase early after midnight, and changes in melatonin concentrations according to gestational age and during the different stages of lactation have been reported. Understudied thus far are the factors impacting on (changes in) melatonin content in human breast milk and their clinical significance in chronobiological adherence in the neonate: maternal as well as environmental aspects have to be investigated in more detail to guide nursing mothers in optimal feeding schedules which probably means a synchronized instead of mistimed feeding practice. This review aims to be thought-provoking regarding the critical role of melatonin in chrononutrition during breastfeeding, highlighting its potential in circadian entrainment and therefore optimizing (neuro)developmental outcomes in the neonatal setting.


Subject(s)
Breast Feeding , Circadian Rhythm , Lactation , Melatonin , Milk, Human , Humans , Melatonin/metabolism , Melatonin/administration & dosage , Milk, Human/chemistry , Milk, Human/metabolism , Circadian Rhythm/physiology , Female , Infant, Newborn , Lactation/physiology , Infant Nutritional Physiological Phenomena/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...