Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Nutrients ; 16(5)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38474876

ABSTRACT

In May of 2022, millions of U.S. parents encountered uncertainty in safely feeding their infants due to the infant formula shortage. METHODS: An anonymous, electronic, cross-sectional, retrospective survey was used. RESULTS: U.S. parents (n = 178) whose infants were ~10 weeks old during the shortage completed the survey. Of parents, 81% switched formulas during the shortage, 87% switched because they could not find the formula they typically used, 34% switched 3-5 times, 29% of parents visited ≥4 stores/24 h and 26% of parents traveled >20 miles/24 h to purchase formula. Use of infant formula increased (p < 0.01); in infants requiring specialty formula, use of intact cow's milk formula increased (p < 0.05) and use of premature infant formulas decreased (p < 0.05). Infants relying on specialty formulas experienced at least one undesirable outcome compared with non-specialty users. Parents used social media, relatives/friends and healthcare providers for support during the shortage, but their helpfulness scores were suboptimal. Parents reported the need for greater infant formula availability, free prenatal lactation education and postpartum lactation support. CONCLUSIONS: Government, regulatory and healthcare policy oversight are needed to protect the infant feeding system, including more commercially available products, access to banked donor milk and lactation support.


Subject(s)
Consumer Behavior , Infant Formula , Infant , Female , Pregnancy , Animals , Cattle , Humans , Cross-Sectional Studies , Retrospective Studies , Breast Feeding , Parents , Diet
2.
Nutrients ; 15(23)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38068830

ABSTRACT

Photo-based dietary assessment is becoming more feasible as artificial intelligence methods improve. However, advancement of these methods for dietary assessment in research settings has been hindered by the lack of an appropriate dataset against which to benchmark algorithm performance. We conducted the Surveying Nutrient Assessment with Photographs of Meals (SNAPMe) study (ClinicalTrials ID: NCT05008653) to pair meal photographs with traditional food records. Participants were recruited nationally, and 110 enrollment meetings were completed via web-based video conferencing. Participants uploaded and annotated their meal photos using a mobile phone app called Bitesnap and completed food records using the Automated Self-Administered 24-h Dietary Assessment Tool (ASA24®) version 2020. Participants included photos before and after eating non-packaged and multi-serving packaged meals, as well as photos of the front and ingredient labels for single-serving packaged foods. The SNAPMe Database (DB) contains 3311 unique food photos linked with 275 ASA24 food records from 95 participants who photographed all foods consumed and recorded food records in parallel for up to 3 study days each. The use of the SNAPMe DB to evaluate ingredient prediction demonstrated that the publicly available algorithms FB Inverse Cooking and Im2Recipe performed poorly, especially for single-ingredient foods and beverages. Correlations between nutrient estimates common to the Bitesnap and ASA24 dietary assessment tools indicated a range in predictive capacity across nutrients (cholesterol, adjusted R2 = 0.85, p < 0.0001; food folate, adjusted R2 = 0.21, p < 0.05). SNAPMe DB is a publicly available benchmark for photo-based dietary assessment in nutrition research. Its demonstrated utility suggested areas of needed improvement, especially the prediction of single-ingredient foods and beverages.


Subject(s)
Artificial Intelligence , Nutrition Assessment , Humans , Benchmarking , Meals , Nutrients , Diet Records , Diet
3.
BMC Pediatr ; 23(1): 320, 2023 06 24.
Article in English | MEDLINE | ID: mdl-37355589

ABSTRACT

BACKGROUND: In May of 2022, parents living in the United States experienced a dramatic infant formula shortage caused by supply chain issues and the recall of several infant formula products over contamination concerns. METHODS: An anonymous, electronic, cross-sectional survey was designed to understand infant feeding practices, parental experience and perceived support during the crisis. RESULTS: Ninety-nine parents that lived in the U.S. and fulfilled study criteria completed the survey. 66% of respondents were female, and 75% of respondents were recipients of the Special Supplemental Nutrition Program for Women Infant Children (WIC). Parental mean age was 30.0 years, and the mean infant age was 26.8 weeks. The number of individuals that used at least one unsafe infant feeding practice increased from 8% before the infant formula shortage to 48.5% during the shortage (p < 0.001). 79% of parents fed their infants U.S. infant formula brands and 39% of parents fed their infants imported infant formula brands before the shortage which were significantly reduced during the shortage to 27% (p < 0.005) and 11% (p < 0.005), respectively. The percentage of parents that reported infant feeding practices before and during the infant formula shortage significantly increased from 2 to 28% for banked donor milk use (p < 0.005); 5-26% for use of human milk from informal sharing (p < 0.005); and 2-29% for use of watered-down infant formula (p < 0.005). The resources that parents reported as most helpful in navigating the crisis differed by parental sex and WIC recipient status and included other parents, friends, and family; lactation consultants; healthcare providers; and WIC. CONCLUSIONS: Our study found that feeding practices in response to the infant formula shortage may pose health risks to infants including nutrition and food insecurity. These data suggest the need for policy changes within regulatory and the healthcare system to provide families with clinical prenatal and postnatal lactation support, access to pasteurized banked donor milk, and access to more commercially available products.


Subject(s)
Breast Feeding , Infant Formula , Pregnancy , Child , Infant , Female , Humans , United States , Adult , Male , Cross-Sectional Studies , Parents , Milk, Human
4.
Am J Clin Nutr ; 117 Suppl 1: S28-S42, 2023 04.
Article in English | MEDLINE | ID: mdl-37173059

ABSTRACT

Human milk is universally recognized as the preferred food for infants during the first 6 mo of life because it provides not only essential and conditionally essential nutrients in necessary amounts but also other biologically active components that are instrumental in protecting, communicating important information to support, and promoting optimal development and growth in infants. Despite decades of research, however, the multifaceted impacts of human milk consumption on infant health are far from understood on a biological or physiological basis. Reasons for this lack of comprehensive knowledge of human milk functions are numerous, including the fact that milk components tend to be studied in isolation, although there is reason to believe that they interact. In addition, milk composition can vary greatly within an individual as well as within and among populations. The objective of this working group within the Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN) Project was to provide an overview of human milk composition, factors impacting its variation, and how its components may function to coordinately nourish, protect, and communicate complex information to the recipient infant. Moreover, we discuss the ways whereby milk components might interact such that the benefits of an intact milk matrix are greater than the sum of its parts. We then apply several examples to illustrate how milk is better thought of as a biological system rather than a more simplistic "mixture" of independent components to synergistically support optimal infant health.


Subject(s)
Breast Feeding , Milk, Human , Female , Infant , Humans , Infant Nutritional Physiological Phenomena
5.
Nutrients ; 14(8)2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35458202

ABSTRACT

The molecular complexity of the carbohydrates consumed by humans has been deceptively oversimplified due to a lack of analytical methods that possess the throughput, sensitivity, and resolution required to provide quantitative structural information. However, such information is becoming an integral part of understanding how specific glycan structures impact health through their interaction with the gut microbiome and host physiology. This work presents a detailed catalogue of the glycans present in complementary foods commonly consumed by toddlers during weaning and foods commonly consumed by American adults. The monosaccharide compositions of over 800 foods from diverse food groups including Fruits, Vegetables, Grain Products, Beans, Peas, Other Legumes, Nuts, Seeds; Sugars, Sweets and Beverages; Animal Products, and more were obtained and used to construct the "Davis Food Glycopedia" (DFG), an open-access database that provides quantitative structural information on the carbohydrates in food. While many foods within the same group possessed similar compositions, hierarchical clustering analysis revealed similarities between different groups as well. Such a Glycopedia can be used to formulate diets rich in specific monosaccharide residues to provide a more targeted modulation of the gut microbiome, thereby opening the door for a new class of prophylactic or therapeutic diets.


Subject(s)
Fabaceae , Food , Animals , Diet , Fruit , Monosaccharides , Polysaccharides , Vegetables
6.
Nutrients ; 14(7)2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35406036

ABSTRACT

Bifidobacterium species are beneficial and dominant members of the breastfed infant gut microbiome; however, their health benefits are partially species-dependent. Here, we characterize the species and subspecies of Bifidobacterium in breastfed infants around the world to consider the potential impact of a historic dietary shift on the disappearance of B. longum subsp. infantis in some populations. Across populations, three distinct patterns of Bifidobacterium colonization emerged: (1) The dominance of Bifidobacterium longum subspecies infantis, (2) prevalent Bifidobacterium of multiple species, and (3) the frequent absence of any Bifidobacterium. These patterns appear related to a country's history of breastfeeding, with infants in countries with historically high rates of long-duration breastfeeding more likely to be colonized by B. longum subspecies infantis compared with infants in countries with histories of shorter-duration breastfeeding. In addition, the timing of infant colonization with B. longum subsp. infantis is consistent with horizontal transmission of this subspecies, rather than the vertical transmission previously reported for other Bifidobacterium species. These findings highlight the need to consider historical and cultural influences on the prevalence of gut commensals and the need to understand epidemiological transmission patterns of Bifidobacterium and other major commensals.


Subject(s)
Bifidobacterium longum , Gastrointestinal Microbiome , Bifidobacterium , Breast Feeding , Cross-Sectional Studies , Female , Humans , Infant
7.
J Nutr ; 152(5): 1239-1253, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35179194

ABSTRACT

BACKGROUND: Human milk oligosaccharides (HMOs) are an abundant class of compounds found in human milk and have been linked to the development of the infant, and specifically the brain, immune system, and gut microbiome. OBJECTIVES: Advanced analytical methods were used to obtain relative quantitation of many structures in approximately 2000 samples from over 1000 mothers in urban, semirural, and rural sites across geographically diverse countries. METHODS: LC-MS-based analytical methods were used to profile the compounds with broad structural coverage and quantitative information. The profiles revealed their structural heterogeneity and their potential biological roles. Comparisons of HMO compositions were made between mothers of different age groups, lactation periods, infant sexes, and residing geographical locations. RESULTS: A common behavior found among all sites was a decrease in HMO abundances during lactation until approximately postnatal month 6, where they remained relatively constant. The greatest variations in structural abundances were associated with the presence of α(1,2)-fucosylated species. Genomic analyses of the mothers were not performed; instead, milk was phenotyped according to the abundances of α(1,2)-fucosylated structures. Mothers from the South American sites tended to have higher proportions of phenotypic secretors [mothers with relatively high concentrations of α(1,2)-fucosylated structures] in their populations compared to the rest of the globe, with Bolivia at ∼100% secretors, Peru at ∼97%, Brazil at ∼90%, and Argentina at ∼85%. Conversely, the cohort sampled in Africa manifested the lowest proportion of secretors (South Africa ∼ 63%, the Gambia ∼ 64%, and Malawi ∼ 75%). Furthermore, we compared total abundances of HMOs in secretors compared with nonsecretors and found that nonsecretors have lower abundances of HMOs compared to secretors, regardless of geographical location. We also observed compositional differences of the 50+ most abundant HMOs between milk types and geographical locations. CONCLUSIONS: This study represents the largest structural HMO study to date and reveals the general behavior of HMOs during lactation among different populations.


Subject(s)
Milk, Human , Oligosaccharides , Breast Feeding , Female , Humans , Infant , Lactation , Malawi , Milk, Human/chemistry , Oligosaccharides/chemistry
8.
Pediatr Res ; 91(3): 627-636, 2022 02.
Article in English | MEDLINE | ID: mdl-33762689

ABSTRACT

BACKGROUND: Recent studies have reported a dysfunctional gut microbiome in breastfed infants. Probiotics have been used in an attempt to restore the gut microbiome; however, colonization has been transient, inconsistent among individuals, or has not positively impacted the host's gut. METHODS: This is a 2-year follow-up study to a randomized controlled trial wherein 7-day-old infants received 1.8 × 1010 colony-forming unit Bifidobacterium longum subsp. infantis (B. infantis) EVC001 (EVC) daily for 21 days or breast milk alone (unsupplemented (UNS)). In the follow-up study, mothers (n = 48) collected infant stool at 4, 6, 8, 10, and 12 months postnatal and completed the health-diet questionnaires. RESULTS: Fecal B. infantis was 2.5-3.5 log units higher at 6-12 months in the EVC group compared with the UNS group (P < 0.01) and this relationship strengthened with the exclusion of infants who consumed infant formula and antibiotics. Infants in the EVC group had significantly higher Bifidobacteriaceae and lower Bacteroidaceae and Lachnospiraceae (P < 0.05). There were no differences in any health conditions between the two groups. CONCLUSIONS: Probiotic supplementation with B. infantis within the first month postnatal, in combination with breast milk, resulted in stable colonization that persisted until at least 1 year postnatal. IMPACT: A dysfunctional gut microbiome in breastfed infants is common in resource-rich nations and associated with an increased risk of immune diseases. Probiotics only transiently exist in the gut without persistent colonization or altering the gut microbiome. This is the first study to show that early probiotic supplementation with B. infantis with breast milk results in stable colonization of B. infantis and improvements to the gut microbiome 1 year postnatal. This study addresses a key gap in the literature whereby probiotics can restore the gut microbiome if biologically selected microorganisms are matched with their specific food in an open ecological niche.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Bifidobacterium longum subspecies infantis , Breast Feeding , Feces/microbiology , Female , Follow-Up Studies , Humans , Infant , Milk, Human
9.
Nutrients ; 13(12)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34959944

ABSTRACT

Streptococcus salivarius (S. salivarius) K12 supplementation has been found to reduce the risk of recurrent upper respiratory tract infections. Yet, studies have not reported the effect of supplementation on oral S. salivarius K12 levels or the salivary microbiome. This clinical trial was designed to determine how supplementation with S. salivarius K12 influences the oral microbiome. In a randomized, double-blind, placebo-controlled trial, 13 healthy adults received a probiotic powder (PRO) containing Lactobacillus acidophilus, Bifidobacterium lactis, and S. salivarius K12 and 12 healthy adults received a placebo-control powder (CON) (n = 12) for 14 consecutive days. Oral S. salivarius K12 and total bacteria were quantified by qPCR and the overall oral microbiome was measured using 16S rRNA amplicon sequencing. Supplementation significantly increased mean salivary S. salivarius K12 levels by 5 logs compared to baseline for the PRO group (p < 0.0005), which returned to baseline 2 weeks post-supplementation. Compared with the CON group, salivary S. salivarius K12 was 5 logs higher in the PRO group at the end of the supplementation period (p < 0.001). Neither time nor supplementation influenced the overall oral microbiome. Supplementation with a probiotic cocktail containing S. salivarius K12 for two weeks significantly increased levels of salivary S. salivarius K12.


Subject(s)
Dietary Supplements , Healthy Volunteers , Probiotics/administration & dosage , Probiotics/pharmacology , Saliva/microbiology , Streptococcus salivarius , Adult , Double-Blind Method , Female , Humans , Male , Recurrence , Respiratory Tract Infections/prevention & control
10.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34143954

ABSTRACT

Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon ß (IFNß) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.


Subject(s)
Bifidobacterium/physiology , Immune System/growth & development , Immune System/microbiology , Anti-Bacterial Agents/pharmacology , Biomarkers/metabolism , Breast Feeding , CD4-Positive T-Lymphocytes/immunology , Cell Polarity , Cell Proliferation , Cytokines/metabolism , Feces/chemistry , Feces/microbiology , Galectin 1/metabolism , Gastrointestinal Microbiome , Humans , Indoles/metabolism , Infant, Newborn , Inflammation/blood , Inflammation/genetics , Intestinal Mucosa/immunology , Metabolome , Milk, Human/chemistry , Oligosaccharides/metabolism , Th17 Cells/immunology , Th2 Cells/immunology , Water
11.
J Agric Food Chem ; 69(23): 6676-6689, 2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34098718

ABSTRACT

Existing methods for the analysis of pesticides in human breast milk involve multiple extraction steps requiring large sample and solvent volumes, which can be a major obstacle in large epidemiologic studies. Here, we developed a simple, low-volume method for extracting organophosphates, pyrethroids, carbamates, atrazine, and imidacloprid from 100 to 200 µL of human breast milk. Multiple extraction protocols were tested including microwave-assisted acid/base digestion and double-solvent extraction with 2 or 20 mL of 2:1 (v/v) dichloromethane/hexane, with or without subsequent solid-phase extraction (SPE) cleanup. Samples were analyzed by liquid chromatography tandem mass spectrometry. Analyte recoveries and reproducibility were highest when 100-200 µL of milk were extracted with 2 mL of dichloromethane/hexane without subsequent SPE steps. Analysis of 79 breast milk samples using this method revealed the presence of carbamates, organophosphates, pyrethroids, and imidacloprid at detection frequencies of 79-96, 53-90, 1-7, and 61%, respectively. This study demonstrates the feasibility of a simple low-volume extraction method for measuring pesticides in human breast milk.


Subject(s)
Pesticides , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Female , Humans , Milk, Human/chemistry , Pesticides/analysis , Reproducibility of Results , Solid Phase Extraction
12.
Nat Commun ; 11(1): 3963, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770134

ABSTRACT

Polysaccharides are the most abundant biomolecules in nature, but are the least understood in terms of their chemical structures and biological functions. Polysaccharides cannot be simply sequenced because they are often highly branched and lack a uniform structure. Furthermore, large polymeric structures cannot be directly analyzed by mass spectrometry techniques, a problem that has been solved for polynucleotides and proteins. While restriction enzymes have advanced genomic analysis, and trypsin has advanced proteomic analysis, there has been no equivalent enzyme for universal polysaccharide digestion. We describe the development and application of a chemical method for producing oligosaccharides from polysaccharides. The released oligosaccharides are characterized by advanced liquid chromatography-mass spectrometry (LC-MS) methods with high sensitivity, accuracy and throughput. The technique is first used to identify polysaccharides by oligosaccharide fingerprinting. Next, the polysaccharide compositions of food and feces are determined, further illustrating the utility of technique in food and clinical studies.


Subject(s)
Oligosaccharides/chemistry , Polysaccharides/metabolism , Bacteria/metabolism , Galactose/analogs & derivatives , Glucans/chemistry , Glucans/metabolism , Humans , Infant , Mannans/chemistry , Mannans/metabolism , Oxidation-Reduction , Polymerization , Time Factors , Xylans/chemistry , Xylans/metabolism
13.
J Nutr Biochem ; 79: 108340, 2020 05.
Article in English | MEDLINE | ID: mdl-32028108

ABSTRACT

Human milk oligosaccharides play a vital role in the development of the gut microbiome in the human infant. Although oligosaccharides derived from bovine milk (BMO) differ in content and profile with those derived from human milk (HMO), several oligosaccharide structures are shared between the species. BMO are commercial alternatives to HMO, but their fate in the digestive tract of healthy adult consumers is unknown. Healthy human subjects consumed two BMO doses over 11-day periods each and provided fecal samples. Metatranscriptomics of fecal samples were conducted to determine microbial and host gene expression in response to the supplement. Fecal samples were also analyzed by mass spectrometry to determine levels of undigested BMO. No changes were observed in microbial gene expression across all participants. Repeated sampling enabled subject-specific analyses: four of six participants had minor, yet statistically significant, changes in microbial gene expression. No significant change was observed in the gene expression of host cells exfoliated in stool. Levels of BMO excreted in feces after supplementation were not significantly different from baseline and were not correlated with dosage or expressed microbial enzyme levels. Collectively, these data suggest that BMO are fully fermented in the human gastrointestinal tract upstream of the distal colon. Additionally, the unaltered host transcriptome provides further evidence for the safety of BMO as a dietary supplement or food ingredient. Further research is needed to investigate potential health benefits of this completely fermentable prebiotic that naturally occurs in cow's milk.


Subject(s)
Feces/chemistry , Gastrointestinal Microbiome/genetics , Milk/chemistry , Oligosaccharides/analysis , Oligosaccharides/genetics , Adolescent , Adult , Animals , Cross-Over Studies , Dietary Supplements , Female , Glycomics , Humans , Male , Milk, Human/chemistry , Transcriptome , Young Adult
14.
Mol Cell Proteomics ; 18(11): 2165-2177, 2019 11.
Article in English | MEDLINE | ID: mdl-31409668

ABSTRACT

Secretory Immunoglobulin A (SIgA) is central to mucosal immunity: represents one of the main immunological mechanisms of defense against the potential attack of pathogens. During lactation SIgA is produced by plasmablasts in the mammary gland and is present in breast milk, playing a vital role in the passive immunity of the newborn. Interestingly, the different components of SIgA are highly N-glycosylated, and these N-Glycans have an essential role in health maintenance. In this work, we performed a glycomic study to compare N-glycosylation of SIgA purified from mature breast milk and saliva, and plasma IgA from the same lactating participants. Our results revealed a greater diversity than previously reported, with 89 glycan compositions that may correspond to over 250 structures. Among these glycans, 54 glycan compositions were characterized as body-fluid specific. Most of these unique N-Glycan compositions identified in SIgA from mature milk and IgA from plasma were fucosylated and both fucosylated and sialylated species, whereas in salivary SIgA the unique structures were mainly undecorated complex N-Glycans. In addition, we evaluated the effect of delivery mode on (S)IgA glycosylation. Lactating participants who had given birth by vaginal delivery presented an increased proportion of high mannose and fucosylated glycans in salivary SIgA, and selected high mannose, fucosylated, sialylated, and both fucosylated and sialylated glycans in plasma IgA, indicating that the hormonal changes during vaginal delivery could affect plasma and saliva IgA. These results reveal the structural details that provide a new dimension to the roles of (S)IgA N-Glycans in different tissues, and especially in maternal and new-born protection and infant development. The design of optimal recombinant IgA molecules specifically targeted to protect mucosal surfaces will need to include this dimension of structural detail.


Subject(s)
Immunoglobulin A, Secretory/analysis , Immunoglobulin A/analysis , Lactation , Milk, Human/metabolism , Plasma/metabolism , Polysaccharides/analysis , Saliva/metabolism , Female , Glycosylation , Humans
15.
Article in English | MEDLINE | ID: mdl-31423298

ABSTRACT

Background: Antibiotic-resistant (AR) bacteria are a global threat. AR bacteria can be acquired in early life and have long-term sequelae. Limiting the spread of antibiotic resistance without triggering the development of additional resistance mechanisms is of immense clinical value. Here, we show how the infant gut microbiome can be modified, resulting in a significant reduction of AR genes (ARGs) and the potentially pathogenic bacteria that harbor them. Methods: The gut microbiome was characterized using shotgun metagenomics of fecal samples from two groups of healthy, term breastfed infants. One group was fed B. infantis EVC001 in addition to receiving lactation support (n = 29, EVC001-fed), while the other received lactation support alone (n = 31, controls). Coliforms were isolated from fecal samples and genome sequenced, as well as tested for minimal inhibitory concentrations against clinically relevant antibiotics. Results: Infants fed B. infantis EVC001 exhibited a change to the gut microbiome, resulting in a 90% lower level of ARGs compared to controls. ARGs that differed significantly between groups were predicted to confer resistance to beta lactams, fluoroquinolones, or multiple drug classes, the majority of which belonged to Escherichia, Clostridium, and Staphylococcus. Minimal inhibitory concentration assays confirmed the resistance phenotypes among isolates with these genes. Notably, we found extended-spectrum beta lactamases among healthy, vaginally delivered breastfed infants who had never been exposed to antibiotics. Conclusions: Colonization of the gut of breastfed infants by a single strain of B. longum subsp. infantis had a profound impact on the fecal metagenome, including a reduction in ARGs. This highlights the importance of developing novel approaches to limit the spread of these genes among clinically relevant bacteria. Future studies are needed to determine whether colonization with B. infantis EVC001 decreases the incidence of AR infections in breastfed infants. Trial registration: This clinical trial was registered at ClinicalTrials.gov, NCT02457338.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/classification , Drug Resistance, Microbial , Metagenomics/methods , Probiotics/administration & dosage , Bacteria/drug effects , Bacteria/isolation & purification , Bifidobacterium longum subspecies infantis/physiology , Breast Feeding , Case-Control Studies , Feces/microbiology , Gastrointestinal Microbiome , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Microbial Sensitivity Tests , Whole Genome Sequencing , beta-Lactamases/genetics
16.
Pediatr Res ; 86(6): 749-757, 2019 12.
Article in English | MEDLINE | ID: mdl-31443102

ABSTRACT

BACKGROUND: Infant gut dysbiosis, often associated with low abundance of bifidobacteria, is linked to impaired immune development and inflammation-a risk factor for increased incidence of several childhood diseases. We investigated the impact of B. infantis EVC001 colonization on enteric inflammation in a subset of exclusively breastfed term infants from a larger clinical study. METHODS: Stool samples (n = 120) were collected from infants randomly selected to receive either 1.8 × 1010 CFU B. infantis EVC001 daily for 21 days (EVC001) or breast milk alone (controls), starting at day 7 postnatal. The fecal microbiome was analyzed using 16S ribosomal RNA, proinflammatory cytokines using multiplexed immunoassay, and fecal calprotectin using ELISA at three time points: days 6 (Baseline), 40, and 60 postnatal. RESULTS: Fecal calprotectin concentration negatively correlated with Bifidobacterium abundance (P < 0.0001; ρ = -0.72), and proinflammatory cytokines correlated with Clostridiaceae and Enterobacteriaceae, yet negatively correlated with Bifidobacteriaceae abundance. Proinflammatory cytokines were significantly lower in EVC001-fed infants on days 40 and 60 postnatally compared to baseline and compared to control infants. CONCLUSION: Our findings indicate that gut dysbiosis (absence of B. infantis) is associated with increased intestinal inflammation. Early addition of EVC001 to diet represents a novel strategy to prevent enteric inflammation during a critical developmental phase.


Subject(s)
Bifidobacterium longum subspecies infantis/growth & development , Breast Feeding , Enteritis/prevention & control , Cytokines/metabolism , Enteritis/metabolism , Enteritis/microbiology , Feces/chemistry , Feces/microbiology , Female , Gastrointestinal Microbiome , Humans , Infant, Newborn , Inflammation Mediators/metabolism , Leukocyte L1 Antigen Complex/analysis , Male , Prospective Studies
17.
J Nutr ; 149(8): 1317-1325, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31098625

ABSTRACT

BACKGROUND: Proteins in human milk are essential and known to support the growth, development, protection, and health of the newborn. These proteins are highly modified by glycans that are currently being recognized as vital to protein structure, stability, function, and health of the intestinal mucosa. Although milk proteins have been studied, the quantitative changes in milk proteins and their respective site-specific glycosylation are unknown. OBJECTIVE: This study expanded the analytical tools for milk proteins and their site-specific glycosylation and applied these tools to a large cohort to determine changes in individual protein concentrations and their site-specific N-glycosylation across lactation. DESIGN: A tandem mass spectrometry method was applied to 231 breast-milk samples from 33 mothers in Davis, California, obtained during 7 different periods of lactation. Dynamic changes in the absolute abundances of milk proteins, as well as variation in site-specific N-glycosylation of individual proteins, were quantified. RESULTS: α-Lactalbumin, ß-casein, k-casein, and α-antitrypsin were significantly increased from colostrum to transitional milk (4.37 ± 1.33 g/L to 6.41 ± 0.72 g/L, 2.25 ± 0.86 g/L to 2.59 ± 0.78 g/L, 1.33 ± 0.44 g/L to 1.60 ± 0.39 g/L, and 0.09 ± 0.10 g/L to 0.11 ± 0.04 g/L, respectively; P < 0.002). α-Lactalbumin (37%), ß-casein (9%), and lysozyme (159%) were higher in mature milk than in colostrum. Glycans exhibited different behavior. Fucosylated glycans of lactoferrin and high-mannose, undecorated, fucosylated, sialylated, and combined fucosylated + sialylated glycans of secretory immunoglobulin A increased during lactation even when the concentrations of the parent proteins decreased. CONCLUSIONS: Proteins in healthy mothers vary dynamically through lactation to support the development of infants. Individual milk proteins carried unique glycan modifications that varied systematically in structure even with site specificity. The role of glycosylation in human milk proteins will be important in understanding the functional components of human milk. This trial was registered at clinicaltrials.gov as NCT01817127.


Subject(s)
Lactation , Milk Proteins/metabolism , Milk, Human/metabolism , Cohort Studies , Colostrum/metabolism , Female , Glycosylation , Humans , Pregnancy , Tandem Mass Spectrometry
18.
BMC Pediatr ; 19(1): 2, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30606146

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is a devastating disease of intestinal inflammation that primarily affects premature infants. A potential risk factor for necrotizing enterocolitis is exposure of the premature neonatal intestine to environmental bacteria and their proinflammatory products such as lipopolysaccharide. The metalloenzyme alkaline phosphatase (ALP) has been shown to reduce lipopolysaccharide-mediated inflammation. Additionally, premature rat pups have reduced alkaline phosphatase activity and expression as compared to full term pups. To explore the possibility that the human premature neonatal intestine has a paucity of alkaline phosphatase activity, we measured endogenously produced intestinal alkaline phosphatase activity in meconium as a function of gestational age. To test whether breast milk could serve as a source of exogenous alkaline phosphatase to the neonatal intestine through ingestion, we measured alkaline phosphatase activity in breast milk across a range of time points post-birth. METHODS: Alkaline phosphatase activity was quantified in 122 meconium samples from infants of gestational ages ranging from 24 to 40 weeks and in 289 breast milk samples collected from 78 individual mothers between days 2-49 post-birth. RESULTS: We observed a strong positive correlation between the meconium alkaline phosphatase activity and gestational age, with preterm infants having lower meconium alkaline phosphatase activities than early term or term infants. Breast milk alkaline phosphatase activity was highest in the first week post-birth, with peak alkaline phosphatase activity at day 2 post-birth, followed by relatively low alkaline phosphatase activity in weeks 2-7. CONCLUSIONS: Our results are consistent with the two major risk factors for necrotizing enterocolitis development, preterm birth and lack of breast milk feeding, both contributing to a paucity of alkaline phosphatase activity and impaired capacity to detoxify proinflammatory bacterial products such as lipopolysaccharide.


Subject(s)
Alkaline Phosphatase/metabolism , Enterocolitis, Necrotizing/etiology , Intestines/enzymology , Milk, Human/enzymology , Alkaline Phosphatase/analysis , Gestational Age , Humans , Infant , Infant, Newborn , Milk, Human/chemistry
19.
PLoS One ; 14(1): e0210064, 2019.
Article in English | MEDLINE | ID: mdl-30625189

ABSTRACT

Over half of all children with autism spectrum disorders (ASD) have gastrointestinal (GI) co-morbidities including chronic constipation, diarrhea, and irritable bowel syndrome. The severity of these symptoms has been correlated with the degree of GI microbial dysbiosis. The study objective was to assess tolerability of a probiotic (Bifidobacterium infantis) in combination with a bovine colostrum product (BCP) as a source of prebiotic oligosaccharides and to evaluate GI, microbiome and immune factors in children with ASD and GI co-morbidities. This pilot study is a randomized, double blind, controlled trial of combination treatment (BCP + B. infantis) vs. BCP alone in a cross-over study in children ages 2-11 with ASD and GI co-morbidities (n = 8). This 12-week study included 5 weeks of probiotic-prebiotic supplementation, followed by a two-week washout period, and 5 weeks of prebiotic only supplementation. The primary outcome of tolerability was assessed using validated questionnaires of GI function and atypical behaviors, along with side effects. Results suggest that the combination treatment is well-tolerated in this cohort. The most common side effect was mild gassiness. Some participants on both treatments saw a reduction in the frequency of certain GI symptoms, as well as reduced occurrence of particular aberrant behaviors. Improvement may be explained by a reduction in IL-13 and TNF-α production in some participants. Although limited conclusions can be drawn from this small pilot study, the results support the need for further research into the efficacy of these treatments.


Subject(s)
Autistic Disorder/drug therapy , Colostrum , Gastrointestinal Diseases/drug therapy , Probiotics/therapeutic use , Animals , Autistic Disorder/physiopathology , Cattle , Child , Child, Preschool , Double-Blind Method , Female , Gastrointestinal Diseases/physiopathology , Humans , Interleukin-13/metabolism , Male , Prebiotics , Tumor Necrosis Factor-alpha/metabolism
20.
J Agric Food Chem ; 66(26): 6851-6859, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29799744

ABSTRACT

Oligosaccharides are known to affect the health of infants. The analysis of these complex molecules in (human) milk samples requires state-of-the-art techniques. This study analyzed the composition and concentration of oligosaccharides in early (day 3) and mature (day 42) human milk as well as in five different infant formula brands. The oligosaccharide content decreased in human milk from 9.15 ± 0.25 g/L at day 3 to 6.38 ± 0.29 g/L at day 42 of lactation. All formulas resulted to be fortified with galacto-oligosaccharides, with one also fortified with polydextrose and another with long-chain fructo-oligosaccharides. About 130 unique oligosaccharide structures were identified in the human milk samples, whereas infant formula contained less diversity of structures. The comparisons indicated that composition and abundance of oligosaccharides unique to human milk are not yet reproduced in infant formulas. The analytical workflow developed is suitable for the determination of prebiotic oligosaccharides in foods that contain diverse carbohydrate structures.


Subject(s)
Infant Formula/chemistry , Milk, Human/chemistry , Oligosaccharides/analysis , Chromatography, High Pressure Liquid , Female , Food, Formulated/analysis , Humans , Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL