Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.493
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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 , Myelin Sheath/metabolism , Mice , Animals, Newborn/growth & development , Milk, Human/chemistry , Milk, Human/metabolism , Humans , Female , Hydrogen Peroxide/metabolism , Brain/metabolism , Brain/growth & development , Reactive Oxygen Species/metabolism , Hippocampus/metabolism , Hippocampus/growth & development , Male , Mice, Inbred C57BL
7.
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
8.
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
9.
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
10.
J Extracell Vesicles ; 13(4): e12422, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38602306

ABSTRACT

Human milk contains extracellular vesicles (HMEVs). Pre-clinical models suggest that HMEVs may enhance intestinal function and limit inflammation; however, it is unknown if HMEVs or their cargo survive neonatal human digestion. This limits the ability to leverage HMEV cargo as additives to infant nutrition or as therapeutics. This study aimed to develop an EV isolation pipeline from small volumes of human milk and neonatal intestinal contents after milk feeding (digesta) to address the hypothesis that HMEVs survive in vivo neonatal digestion to be taken up intestinal epithelial cells (IECs). Digesta was collected from nasoduodenal sampling tubes or ostomies. EVs were isolated from raw and pasteurized human milk and digesta by density-gradient ultracentrifugation following two-step skimming, acid precipitation of caseins, and multi-step filtration. EVs were validated by electron microscopy, western blotting, nanoparticle tracking analysis, resistive pulse sensing, and super-resolution microscopy. EV uptake was tested in human neonatal enteroids. HMEVs and digesta EVs (dEVs) show typical EV morphology and are enriched in CD81 and CD9, but depleted of ß-casein and lactalbumin. HMEV and some dEV fractions contain mammary gland-derived protein BTN1A1. Neonatal human enteroids rapidly take up dEVs in part via clathrin-mediated endocytosis. Our data suggest that EVs can be isolated from digestive fluid and that these dEVs can be absorbed by IECs.


Subject(s)
Body Fluids , Extracellular Vesicles , Infant, Newborn , Infant , Humans , Milk, Human/metabolism , Extracellular Vesicles/metabolism , Caseins/metabolism
11.
Thromb Res ; 237: 141-144, 2024 May.
Article in English | MEDLINE | ID: mdl-38593524

ABSTRACT

The use of direct oral anticoagulants (DOACs) in breastfeeding women is currently challenging due to limited safety data for breastfeeding infants, and there have been no previous studies on the drug concentration in breastfeeding infants. We treated 2 patients (one case was twin pregnancy) with venous thromboembolisms in breastfeeding women administered rivaroxaban at our institution. Blood samples from the mothers and breastmilk samples were collected at time 0 and 2 h after the rivaroxaban administration, breastfeeding was conducted 2 h after the rivaroxaban administration, and blood samples from the infants were collected 2 h after breastfeeding (4 h after maternal rivaroxaban administration). The milk-to-plasma (M:P) ratios were 0.27 in Case 1 and 0.32 in Case 2. The estimated relative infant dose (RID) was 0.82 % in Case 1 Children 1 and 2, and 1.27 % in Case 2. The rivaroxaban concentration in the infant plasma was below the lower limit of quantification in all infants. In addition, even in the high-exposure case simulation based on 5 days of breastfeeding in Case 2, the infant plasma concentration level was below the lower limit of quantification. At 3 months of follow-up, breastfeeding was continued, and all infants grew and developed without any health problems including bleeding events. The current case series showed that there were no pharmacokinetic or clinical concerns for breastfeeding women or breastfed infants, and provides support for rivaroxaban as a safe treatment option for these patients.


Subject(s)
Breast Feeding , Factor Xa Inhibitors , Milk, Human , Rivaroxaban , Humans , Rivaroxaban/therapeutic use , Rivaroxaban/pharmacokinetics , Female , Adult , Factor Xa Inhibitors/therapeutic use , Factor Xa Inhibitors/pharmacokinetics , Milk, Human/chemistry , Milk, Human/metabolism , Infant , Venous Thromboembolism/drug therapy , Infant, Newborn , Pregnancy
12.
J Nutr Sci Vitaminol (Tokyo) ; 70(2): 158-163, 2024.
Article in English | MEDLINE | ID: mdl-38684386

ABSTRACT

The Ussing chamber is a tool for analyzing drug absorption. We investigated whether the Ussing chamber can be used to analyze the process from digestion to absorption of protein in the gastrointestinal tract. Mixtures containing infant formula, whole cow's milk, processed soy milk, enteral nutrition, or human breast milk, were placed in the apical membrane side equipped with Caco-2 cells. After the addition of first pepsin then pancreatin, samples from the apical and basal membranes were collected. Infant formula showed the highest digestibility and absorption rate. This may be attributed to the presence of whey protein, which is rapidly digested and absorbed. The digestion and absorption of human breast milk showed different results in each donor, suggesting that digestion and absorption may vary among individuals. We concluded that the Ussing chamber can continuously analyze the process from digestion to absorption of proteins in the gastrointestinal tract.


Subject(s)
Digestion , Gastrointestinal Tract , Infant Formula , Intestinal Absorption , Milk Proteins , Milk, Human , Milk , Whey Proteins , Digestion/physiology , Humans , Caco-2 Cells , Gastrointestinal Tract/metabolism , Milk, Human/chemistry , Milk, Human/metabolism , Infant Formula/chemistry , Animals , Milk Proteins/metabolism , Milk/chemistry , Dietary Proteins/metabolism , Dietary Proteins/pharmacokinetics , Enteral Nutrition/methods , Soy Milk/chemistry , Infant , Pepsin A/metabolism
13.
Arch Pharm Res ; 47(4): 325-340, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38561494

ABSTRACT

Sialyllactoses (SLs) primarily include sialylated human milk oligosaccharides (HMOs) and bovine milk oligosaccharides (BMOs). First, the safety assessment of 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL) revealed low toxicity in various animal models and human participants. SLs constitute a unique milk component, highlighting the essential nutrients and bioactive components crucial for infant development, along with numerous associated health benefits for various diseases. This review explores the safety, biosynthesis, and potential biological effects of SLs, with a specific focus on their influence across various physiological systems, including the gastrointestinal system, immune disorders, rare genetic disorders (such as GNE myopathy), cancers, neurological disorders, cardiovascular diseases, diverse cancers, and viral infections, thus indicating their therapeutic potential.


Subject(s)
Lactose/analogs & derivatives , Milk, Human , Milk , Oligosaccharides , Humans , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Animals , Milk/chemistry , Milk, Human/chemistry , Milk, Human/metabolism , Cattle
14.
JPEN J Parenter Enteral Nutr ; 48(4): 479-485, 2024 May.
Article in English | MEDLINE | ID: mdl-38566550

ABSTRACT

BACKGROUND: Extracellular vesicles in human milk are critical in supporting newborn growth and development. Bioavailability of dietary extracellular vesicles may depend on the composition of membrane lipids. Single-nucleotide polymorphisms (SNPs) in the fatty acid desaturase gene cluster impact the content of long-chain polyunsaturated fatty acids in human milk phospholipids. This study investigated the relation between variation in FADS1 and FADS2 with the content of polyunsaturated fatty acids in extracellular vesicles from human milk. METHODS: Milk was obtained from a cohort of mothers (N = 70) at 2-4 weeks of lactation. SNPs in the FADS gene locus were determined using pyrosequencing for rs174546 in FADS1 and rs174575 in FADS2. Quantitative lipidomic analysis of polyunsaturated fatty acids in human milk and extracellular vesicles from human milk was completed by gas chromatography-mass spectrometry. RESULTS: The rs174546 and rs174575 genotypes were independent predictors of the arachidonic acid content in extracellular vesicles. The rs174546 genotype also predicted eicosapentaenoic acid and docosahexaenoic acid in extracellular vesicles. The reduced content of long-chain polyunsaturated fatty acids in extracellular vesicles in human milk may be due to lower fatty acid desaturase activity in mothers who are carriers of the A allele in rs174546 or the G allele in rs174575. CONCLUSION: The polyunsaturated fatty acid composition of milk extracellular vesicles is predicted by the FADS genotype. These findings yield novel insights regarding extracellular vesicle content and composition that can inform the design of future research to explore how lipid metabolites impact the bioavailability of human milk extracellular vesicles.


Subject(s)
Delta-5 Fatty Acid Desaturase , Extracellular Vesicles , Fatty Acid Desaturases , Fatty Acids, Unsaturated , Genotype , Milk, Human , Polymorphism, Single Nucleotide , Humans , Milk, Human/chemistry , Milk, Human/metabolism , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Female , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Fatty Acids, Unsaturated/analysis , Fatty Acids, Unsaturated/metabolism , Adult , Genetic Association Studies , Cohort Studies , Lactation/genetics , Lactation/metabolism , Docosahexaenoic Acids/analysis , Docosahexaenoic Acids/metabolism
15.
Nutrients ; 16(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542769

ABSTRACT

INTRODUCTION: Breastfeeding is one of the strategies that has been shown to be effective in preventing severe forms of bronchopulmonary dysplasia (BPD). When mother's own milk (MOM) is not available, pasteurized donor milk (DM) is the best alternative. However, the evidence is inconclusive on the difference in the incidence of bronchopulmonary dysplasia (BPD) between patients fed MOM and those fed with DM. As standard DM is usually mature pooled milk donated by mothers who have delivered their babies at term, the potential benefits of preterm milk may be lost. MATERIALS AND METHODS: An observational, retrospective, single-center study was conducted in the neonatology department of a high-complexity hospital. The study included newborns <32 weeks of gestational age born between January 2020 and December 2022. When supplemental milk was needed, non-pooled preterm pasteurized donor milk (PDM) matched for gestational age and moment of lactation was used in this study, classifying preterm infants in two groups: mainly MOM (>50% of the milk) or mainly PDM (>50% of the milk). Two groups were established: those who received >50% MOM and those who received >50% PDM. They were also classified according to the diagnosis of DBP: one group included no BPD or grade 1 BPD (noBPD/1), while the other included grade 2 or 3 BPD (BPD 2-3). The objectives of this study were, firstly, to evaluate the incidence of BPD 2-3 among patients who predominantly received PDM versus MOM. Secondly, to analyze differences in the type of human milk received and its nutritional components, as well as to study the growth in patients with or without BPD. RESULTS: One hundred ninety-nine patients were included in the study. A comparison of noBPD/1 versus BPD 2-3 groups between those receiving mainly MOM versus PDM showed no significant differences (19% vs. 20%, p 0.95). PDM colostrum in BPD 2-3 compared to noBPD/1 was higher in protein content (2.24 g/100 mL (SD 0.37) vs. 2.02 g/100 mL (SD 0.29) p < 0.01), although the statistical significance decreased after adjustment for gestational age and birth weight z-score (OR 3.53 (0.86-14.51)). No differences were found in the macronutrients in the mature milk of patients feeding more than 50% PDM in both study groups. Growth of BPD 2-3 showed a greater decrease in the difference in z-scores for height at birth and at discharge compared to noBPD/1 (-1.64 vs. -0.43, p 0.03). CONCLUSIONS: The use of mainly MOM or PDM demonstrates a similar incidence of noBPD/1 or BPD 2-3. Non-pooled and matched by gestational age and time of lactation preterm donor milk can probably be an alternative when mother's own milk is not available, with a similar protective effect in the prevention of severe BPD.


Subject(s)
Bronchopulmonary Dysplasia , Premature Birth , Infant , Female , Infant, Newborn , Humans , Infant, Premature , Bronchopulmonary Dysplasia/epidemiology , Bronchopulmonary Dysplasia/prevention & control , Infant, Very Low Birth Weight , Retrospective Studies , Milk, Human/metabolism , Breast Feeding
16.
Sci Rep ; 14(1): 7401, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38548847

ABSTRACT

Lipids play a critical role in neonate development and breastmilk is the newborn's major source of lipids. Milk lipids directly influence the neonate plasma lipid profile. The milk lipidome is dynamic, influenced by maternal factors and related to the maternal plasma lipidome. The close inter-relationship between the maternal plasma, milk and neonate plasma lipidomes is critical to understanding maternal-child health and nutrition. In this exploratory study, lipidomes of blood and breast milk from Suffolk sheep and matched lamb blood (n = 13), were profiled on day 34 post birth by untargeted mass spectrometry. Comparative multivariate analysis of the three matrices identified distinct differences in lipids and class of lipids amongst them. Paired analysis identified 346 differential lipids (DL) and 31 correlated lipids (CL) in maternal plasma and milk, 340 DL and 32 CL in lamb plasma and milk and 295 DL and 16 CL in maternal plasma and lamb plasma. Conversion of phosphatidic acid to phosphatidyl inositol was the most active pathway in lamb plasma compared to maternal plasma. This exploratory study illustrates the partitioning of lipids across maternal plasma, milk and lamb plasma and the dynamic relationship between them, reiterating the need to study these three matrices as one biological system.


Subject(s)
Lipidomics , Milk , Female , Animals , Sheep , Humans , Milk/metabolism , Milk, Human/metabolism , Nutritional Status , Plasma , Lipids
17.
Food Funct ; 15(7): 3259-3273, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38469864

ABSTRACT

Long-chain polyunsaturated fatty acids (LC-PUFAs), arachidonic acid (ARA, 20:4n-6) and docosahexaenoic acid (DHA, 22:6n-3) are essential in the development of infants. ARA and DHA from breast milk or infant formula are the main sources of access for infants to meet their physiological and metabolic needs. The ratio of ARA to DHA in breast milk varies among regions and different lactation stages. Different ratios of ARA and DHA mainly from algal oil, animal fat, fish oil, and microbial oil, are added to infant formula in different regions and infant age ranges. Supplementing with appropriate ratios of ARA and DHA during infancy promotes brain, neural, visual, and other development aspects. In this review, we first introduced the current intake status of ARA and DHA in different locations, lactation stages, and age ranges in breast milk and infant formula. Finally, we discussed the effect of different ratios of ARA and DHA on infant development. This review provided a comprehensive research basis for the nutritional research of infants who consume different ratios of ARA and DHA.


Subject(s)
Child Development , Docosahexaenoic Acids , Infant , Animals , Female , Child , Humans , Docosahexaenoic Acids/metabolism , Milk, Human/metabolism , Fatty Acids/metabolism , Infant Formula , Eating
18.
J Agric Food Chem ; 72(12): 6213-6225, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38501388

ABSTRACT

Human milk is naturally rich in medium- and long-chain triacylglycerols (MLCT), accounting for approximately 30% of the total fat. However, infant formula fat is prepared using a physical blend of vegetable oils, which rarely contains MLCT, similar to human milk. The differences in MLCT between human milk and infant formulas may cause different lipid metabolisms and physiological effects on infants, which are unknown. This study aimed to analyze the metabolic characteristics of formula lipid containing novel human milk fat substitutes based on MLCT (FL-MLCT) and compare their effects with those of the physical blend of vegetable oils (FL-PB) on lipid metabolism and gut microbiota in mice. Compared with the FL-PB group, the FL-MLCT group showed increased energy expenditure, decreased serum triacylglycerol level, and significantly lower aspartate aminotransferase level, epididymal and perirenal fat weight, and adipocyte size. Moreover, the abundances of Firmicutes/Bacteroidota, Actinobacteriota, and Desulfovibrionaceae were significantly decreased in the FL-MLCT group. Novel human milk fat substitutes MLCT could inhibit visceral fat accumulation, improve liver function, and modulate the mice gut microbiota composition, which may contribute to controlling obesity.


Subject(s)
Fat Substitutes , Gastrointestinal Microbiome , Infant , Humans , Mice , Animals , Triglycerides/metabolism , Lipid Metabolism , Milk, Human/metabolism , Mice, Inbred C57BL , Plant Oils/metabolism , Thermogenesis
19.
J Agric Food Chem ; 72(11): 5860-5866, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38452259

ABSTRACT

Lacto-N-biose I (LNB), termed a Type 1 disaccharide, is an important building block of human milk oligosaccharides. It shows promising prebiotic activity by stimulating the proliferation of many gut-associated bifidobacteria and thus displays good potential in infant foods or supplements. Enzymatic and microbial approaches to LNB synthesis have been studied, almost all of which involve glycosylation of LNB phosphorylase as the final step. Herein, we report a new and easier microbial LNB synthesis strategy through the route "lactose → lacto-N-triose II (LNTri II) → lacto-N-tetraose (LNT) → LNB". A previously constructed LNT-producing Escherichia coli BL21(DE3) strain was engineered for LNB biosynthesis by introducing Bifidobacterium bifidum LnbB. LNB was efficiently produced, accompanied by lactose regeneration. Genomic integration of key pathway genes related to LNTri II and LNT synthesis was performed to enhance LNB titers. The final engineered strain produced 3.54 and 26.88 g/L LNB by shake-flask and fed-batch cultivation, respectively.


Subject(s)
Acetylglucosamine/analogs & derivatives , Escherichia coli , Milk, Human , Infant , Humans , Milk, Human/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Lactose/metabolism , Oligosaccharides/metabolism
20.
Front Immunol ; 15: 1271926, 2024.
Article in English | MEDLINE | ID: mdl-38426086

ABSTRACT

Natural components of breast milk, human milk oligosaccharides (HMOs) and osteopontin (OPN) have been shown to have a variety of functional activities and are widely used in infant formulas. However, the preventive and therapeutic effects of both on influenza viruses are not known. In this study, antiviral assays using a human laryngeal carcinoma cell line (HEP-2) showed that 3'-sialyllactose (3'-SL) and OPN had the best antiviral ability with IC50 values of 33.46 µM and 1.65 µM, respectively. 3'-SL (10 µM) and OPN (4 µM) were used in combination to achieve 75% inhibition. Further studies found that the combination of 200 µg/mL of 3'-SL with 500 µg/mL of OPN exerted the best antiviral ability. The reason for this was related to reduced levels of the cytokines TNF-α, IL-6, and iNOS in relation to mRNA expression. Plaque assay and TCID50 assay found the same results and verified synergistic effects. Our research indicates that a combination of 3'-SL and OPN can effectively reduce inflammatory storms and exhibit anti-influenza virus effects through synergistic action.


Subject(s)
Influenza, Human , Orthomyxoviridae , Infant , Female , Humans , Osteopontin/genetics , Influenza, Human/drug therapy , Milk, Human/metabolism , Oligosaccharides/pharmacology , Antiviral Agents
SELECTION OF CITATIONS
SEARCH DETAIL
...