Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23.859
Filter
Add more filters

Coleção CLAP
Publication year range
1.
Cell ; 187(3): 750-763.e20, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38242132

ABSTRACT

Breastfeeding offers demonstrable benefits to newborns and infants by providing nourishment and immune protection and by shaping the gut commensal microbiota. Although it has been appreciated for decades that breast milk contains complement components, the physiological relevance of complement in breast milk remains undefined. Here, we demonstrate that weanling mice fostered by complement-deficient dams rapidly succumb when exposed to murine pathogen Citrobacter rodentium (CR), whereas pups fostered on complement-containing milk from wild-type dams can tolerate CR challenge. The complement components in breast milk were shown to directly lyse specific members of gram-positive gut commensal microbiota via a C1-dependent, antibody-independent mechanism, resulting in the deposition of the membrane attack complex and subsequent bacterial lysis. By selectively eliminating members of the commensal gut community, complement components from breast milk shape neonate and infant gut microbial composition to be protective against environmental pathogens such as CR.


Subject(s)
Complement System Proteins , Gastrointestinal Microbiome , Milk , Animals , Female , Humans , Infant , Mice , Bacteria , Breast Feeding , Citrobacter rodentium , Complement System Proteins/analysis , Immunologic Factors , Infant Health , Milk, Human , Milk/chemistry , Enterobacteriaceae Infections/immunology
2.
Cell ; 185(23): 4280-4297.e12, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36323316

ABSTRACT

The gut microbiome has an important role in infant health and development. We characterized the fecal microbiome and metabolome of 222 young children in Dhaka, Bangladesh during the first two years of life. A distinct Bifidobacterium longum clade expanded with introduction of solid foods and harbored enzymes for utilizing both breast milk and solid food substrates. The clade was highly prevalent in Bangladesh, present globally (at lower prevalence), and correlated with many other gut taxa and metabolites, indicating an important role in gut ecology. We also found that the B. longum clades and associated metabolites were implicated in childhood diarrhea and early growth, including positive associations between growth measures and B. longum subsp. infantis, indolelactate and N-acetylglutamate. Our data demonstrate geographic, cultural, seasonal, and ecological heterogeneity that should be accounted for when identifying microbiome factors implicated in and potentially benefiting infant development.


Subject(s)
Bifidobacterium longum , Infant , Child , Female , Humans , Child, Preschool , Bifidobacterium longum/metabolism , Bifidobacterium/metabolism , Weaning , Oligosaccharides/metabolism , Bangladesh , Milk, Human , Feces/microbiology
3.
Cell ; 184(6): 1486-1499, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740451

ABSTRACT

Neonates are born with an immature immune system and rely on the transfer of immunity from their mothers. Maternal antibodies are transferred via the placenta and breast milk. Although the role of placentally transferred immunoglobulin G (IgG) is established, less is known about the selection of antibodies transferred via breast milk and the mechanisms by which they provide protection against neonatal disease. Evidence suggests that breast milk antibodies play multifaceted roles, preventing infection and supporting the selection of commensals and tolerizing immunity during infancy. Here, we discuss emerging data related to the importance of breast milk antibodies in neonatal immunity and development.


Subject(s)
Antibodies/metabolism , Milk, Human/immunology , Animals , Homeostasis , Humans , Immunity , Immunologic Factors/pharmacology , Microbiota
4.
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
5.
Nat Immunol ; 24(7): 1098-1109, 2023 07.
Article in English | MEDLINE | ID: mdl-37337103

ABSTRACT

Macrophages are involved in immune defense, organogenesis and tissue homeostasis. Macrophages contribute to the different phases of mammary gland remodeling during development, pregnancy and involution postlactation. Less is known about the dynamics of mammary gland macrophages in the lactation stage. Here, we describe a macrophage population present during lactation in mice. By multiparameter flow cytometry and single-cell RNA sequencing, we identified a lactation-induced CD11c+CX3CR1+Dectin-1+ macrophage population (liMac) that was distinct from the two resident F4/80hi and F4/80lo macrophage subsets present pregestationally. LiMacs were predominantly monocyte-derived and expanded by proliferation in situ concomitant with nursing. LiMacs developed independently of IL-34, but required CSF-1 signaling and were partly microbiota-dependent. Locally, they resided adjacent to the basal cells of the alveoli and extravasated into the milk. We found several macrophage subsets in human milk that resembled liMacs. Collectively, these findings reveal the emergence of unique macrophages in the mammary gland and milk during lactation.


Subject(s)
Lactation , Milk, Human , Pregnancy , Female , Mice , Humans , Animals , Macrophages , Mammary Glands, Animal
6.
Cell ; 181(6): 1202-1204, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32497500

ABSTRACT

ROR-γt+ regulatory T cells (Tregs) of the colon can prevent excessive inflammation but also delay pathogen clearance. How these cells are regulated has remained elusive. In this issue of Cell, Ramanan et al. find that the set-point for ROR-γt+ Tregs is non-genetically maternally inherited during a critical time window after birth through immunoglobulin A present in breast milk.


Subject(s)
Gastrointestinal Microbiome , T-Lymphocytes, Regulatory , Female , Humans , Milk, Human , Nuclear Receptor Subfamily 1, Group F, Member 3 , Th17 Cells
7.
Cell ; 165(4): 842-53, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27133167

ABSTRACT

According to the hygiene hypothesis, the increasing incidence of autoimmune diseases in western countries may be explained by changes in early microbial exposure, leading to altered immune maturation. We followed gut microbiome development from birth until age three in 222 infants in Northern Europe, where early-onset autoimmune diseases are common in Finland and Estonia but are less prevalent in Russia. We found that Bacteroides species are lowly abundant in Russians but dominate in Finnish and Estonian infants. Therefore, their lipopolysaccharide (LPS) exposures arose primarily from Bacteroides rather than from Escherichia coli, which is a potent innate immune activator. We show that Bacteroides LPS is structurally distinct from E. coli LPS and inhibits innate immune signaling and endotoxin tolerance; furthermore, unlike LPS from E. coli, B. dorei LPS does not decrease incidence of autoimmune diabetes in non-obese diabetic mice. Early colonization by immunologically silencing microbiota may thus preclude aspects of immune education.


Subject(s)
Bacteroides/immunology , Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome , Lipopolysaccharides/immunology , Animals , Estonia , Feces/microbiology , Finland , Food Microbiology , Humans , Infant , Mice , Mice, Inbred NOD , Milk, Human/immunology , Russia
8.
Cell ; 165(4): 827-41, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27153495

ABSTRACT

To maintain a symbiotic relationship between the host and its resident intestinal microbiota, appropriate mucosal T cell responses to commensal antigens must be established. Mice acquire both IgG and IgA maternally; the former has primarily been implicated in passive immunity to pathogens while the latter mediates host-commensal mutualism. Here, we report the surprising observation that mice generate T cell-independent and largely Toll-like receptor (TLR)-dependent IgG2b and IgG3 antibody responses against their gut microbiota. We demonstrate that maternal acquisition of these antibodies dampens mucosal T follicular helper responses and subsequent germinal center B cell responses following birth. This work reveals a feedback loop whereby T cell-independent, TLR-dependent antibodies limit mucosal adaptive immune responses to newly acquired commensal antigens and uncovers a broader function for maternal IgG.


Subject(s)
Animals, Newborn/immunology , Gastrointestinal Microbiome , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Milk, Human/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Animals, Newborn/microbiology , B-Lymphocytes/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Signal Transduction , Specific Pathogen-Free Organisms , Toll-Like Receptors/immunology
9.
Cell ; 164(5): 859-71, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26898329

ABSTRACT

Identifying interventions that more effectively promote healthy growth of children with undernutrition is a pressing global health goal. Analysis of human milk oligosaccharides (HMOs) from 6-month-postpartum mothers in two Malawian birth cohorts revealed that sialylated HMOs are significantly less abundant in those with severely stunted infants. To explore this association, we colonized young germ-free mice with a consortium of bacterial strains cultured from the fecal microbiota of a 6-month-old stunted Malawian infant and fed recipient animals a prototypic Malawian diet with or without purified sialylated bovine milk oligosaccharides (S-BMO). S-BMO produced a microbiota-dependent augmentation of lean body mass gain, changed bone morphology, and altered liver, muscle, and brain metabolism in ways indicative of a greater ability to utilize nutrients for anabolism. These effects were also documented in gnotobiotic piglets using the same consortium and Malawian diet. These preclinical models indicate a causal, microbiota-dependent relationship between S-BMO and growth promotion.


Subject(s)
Child Development , Malnutrition/diet therapy , Milk, Human/chemistry , Milk/chemistry , Oligosaccharides/metabolism , Animals , Bacteroides fragilis/genetics , Bifidobacterium/classification , Bifidobacterium/genetics , Brain Chemistry , Disease Models, Animal , Escherichia coli/genetics , Feces/microbiology , Germ-Free Life , Humans , Infant , Malawi , Male , Metabolomics , Mice , Mice, Inbred C57BL , Microbiota
11.
Immunity ; 54(8): 1633-1635, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34380062

ABSTRACT

Immune-system maturation starts early in life, but studies investigating immune-system education in human infants remain scarce. In a recent issue of Cell, Henrick et al. study early gut microbiota and immune-system development in two infant cohorts. The authors describe that Bifidobacteria can use milk sugars to produce immunoregulatory compounds that induce immune tolerance and reduce intestinal inflammation.


Subject(s)
Bifidobacterium/metabolism , Immune System/growth & development , Intestines/immunology , Intestines/microbiology , Milk, Human/chemistry , Oligosaccharides/metabolism , Animals , Breast Feeding , Gastrointestinal Microbiome/physiology , Humans , Immune Tolerance/immunology , Immunologic Factors/chemistry , Immunomodulation/immunology , Infant , Sweden , United States
12.
Nature ; 618(7964): 365-373, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225978

ABSTRACT

Birth presents a metabolic challenge to cardiomyocytes as they reshape fuel preference from glucose to fatty acids for postnatal energy production1,2. This adaptation is triggered in part by post-partum environmental changes3, but the molecules orchestrating cardiomyocyte maturation remain unknown. Here we show that this transition is coordinated by maternally supplied γ-linolenic acid (GLA), an 18:3 omega-6 fatty acid enriched in the maternal milk. GLA binds and activates retinoid X receptors4 (RXRs), ligand-regulated transcription factors that are expressed in cardiomyocytes from embryonic stages. Multifaceted genome-wide analysis revealed that the lack of RXR in embryonic cardiomyocytes caused an aberrant chromatin landscape that prevented the induction of an RXR-dependent gene expression signature controlling mitochondrial fatty acid homeostasis. The ensuing defective metabolic transition featured blunted mitochondrial lipid-derived energy production and enhanced glucose consumption, leading to perinatal cardiac dysfunction and death. Finally, GLA supplementation induced RXR-dependent expression of the mitochondrial fatty acid homeostasis signature in cardiomyocytes, both in vitro and in vivo. Thus, our study identifies the GLA-RXR axis as a key transcriptional regulatory mechanism underlying the maternal control of perinatal cardiac metabolism.


Subject(s)
Fatty Acids , Glucose , Heart , Milk, Human , gamma-Linolenic Acid , Female , Humans , Infant, Newborn , Pregnancy , Chromatin/genetics , Fatty Acids/metabolism , gamma-Linolenic Acid/metabolism , gamma-Linolenic Acid/pharmacology , Gene Expression Regulation/drug effects , Glucose/metabolism , Heart/drug effects , Heart/embryology , Heart/growth & development , Homeostasis , In Vitro Techniques , Milk, Human/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Retinoid X Receptors/metabolism , Transcription Factors/metabolism
13.
N Engl J Med ; 390(16): 1493-1504, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38657245

ABSTRACT

BACKGROUND: Most moderate-to-late-preterm infants need nutritional support until they are feeding exclusively on their mother's breast milk. Evidence to guide nutrition strategies for these infants is lacking. METHODS: We conducted a multicenter, factorial, randomized trial involving infants born at 32 weeks 0 days' to 35 weeks 6 days' gestation who had intravenous access and whose mothers intended to breast-feed. Each infant was assigned to three interventions or their comparators: intravenous amino acid solution (parenteral nutrition) or dextrose solution until full feeding with milk was established; milk supplement given when maternal milk was insufficient or mother's breast milk exclusively with no supplementation; and taste and smell exposure before gastric-tube feeding or no taste and smell exposure. The primary outcome for the parenteral nutrition and the milk supplement interventions was the body-fat percentage at 4 months of corrected gestational age, and the primary outcome for the taste and smell intervention was the time to full enteral feeding (150 ml per kilogram of body weight per day or exclusive breast-feeding). RESULTS: A total of 532 infants (291 boys [55%]) were included in the trial. The mean (±SD) body-fat percentage at 4 months was similar among the infants who received parenteral nutrition and those who received dextrose solution (26.0±5.4% vs. 26.2±5.2%; adjusted mean difference, -0.20; 95% confidence interval [CI], -1.32 to 0.92; P = 0.72) and among the infants who received milk supplement and those who received mother's breast milk exclusively (26.3±5.3% vs. 25.8±5.4%; adjusted mean difference, 0.65; 95% CI, -0.45 to 1.74; P = 0.25). The time to full enteral feeding was similar among the infants who were exposed to taste and smell and those who were not (5.8±1.5 vs. 5.7±1.9 days; P = 0.59). Secondary outcomes were similar across interventions. Serious adverse events occurred in one infant. CONCLUSIONS: This trial of routine nutrition interventions to support moderate-to-late-preterm infants until full nutrition with mother's breast milk was possible did not show any effects on the time to full enteral feeding or on body composition at 4 months of corrected gestational age. (Funded by the Health Research Council of New Zealand and others; DIAMOND Australian New Zealand Clinical Trials Registry number, ACTRN12616001199404.).


Subject(s)
Breast Feeding , Enteral Nutrition , Infant, Premature , Parenteral Nutrition , Female , Humans , Infant , Infant, Newborn , Male , Amino Acids/administration & dosage , Gestational Age , Glucose/administration & dosage , Milk, Human , Smell , Taste , Nutritional Support , Parenteral Nutrition Solutions/therapeutic use , Adiposity
14.
Nat Rev Neurosci ; 22(7): 423-438, 2021 07.
Article in English | MEDLINE | ID: mdl-34021274

ABSTRACT

Recent years have been transformational in regard to the perception of the health risks and benefits of cannabis with increased acceptance of use. This has unintended neurodevelopmental implications given the increased use of cannabis and the potent levels of Δ9-tetrahydrocannabinol today being consumed by pregnant women, young mothers and teens. In this Review, we provide an overview of the neurobiological effects of cannabinoid exposure during prenatal/perinatal and adolescent periods, in which the endogenous cannabinoid system plays a fundamental role in neurodevelopmental processes. We highlight impaired synaptic plasticity as characteristic of developmental exposure and the important contribution of epigenetic reprogramming that maintains the long-term impact into adulthood and across generations. Such epigenetic influence by its very nature being highly responsive to the environment also provides the potential to diminish neural perturbations associated with developmental cannabis exposure.


Subject(s)
Brain/drug effects , Cannabis , Prenatal Exposure Delayed Effects , Adolescent , Adult , Age Factors , Animals , Brain/embryology , Brain/growth & development , Cannabis/adverse effects , Child , Child, Preschool , Dronabinol/adverse effects , Dronabinol/pharmacokinetics , Dronabinol/pharmacology , Endocannabinoids/physiology , Epigenesis, Genetic/drug effects , Female , Humans , Infant , Lactation , Lipase/physiology , Male , Marijuana Smoking , Maternal Exposure , Mice , Milk, Human/chemistry , Neurodevelopmental Disorders/chemically induced , Neuronal Plasticity/drug effects , Neurotransmitter Agents/physiology , Paternal Exposure , Pregnancy , Rats , Receptor, Cannabinoid, CB1/physiology , Species Specificity , Young Adult
15.
Trends Immunol ; 44(8): 644-661, 2023 08.
Article in English | MEDLINE | ID: mdl-37438187

ABSTRACT

Childhood allergy, including asthma, eczema, and food allergies, is a major global health burden, with prevalence increasing dramatically and novel interventions needed. Emerging research suggests that human milk oligosaccharides (HMOs), complex glycans found in breastmilk, have allergy-protective properties, indicating exciting therapeutic potential. This review evaluates current literature on the role of HMOs in allergy, assesses underlying immunological mechanisms, and discusses future research needed to translate findings into clinical implications. HMOs may mediate allergy risk through multiple structure-specific mechanisms, including microbiome modification, intestinal barrier maturation, immunomodulation, and gene regulation. Findings emphasize the importance of breastfeeding encouragement and HMO-supplemented formula milk for high allergy-risk infants. Although further investigation is necessary to determine the most efficacious structures against varying allergy phenotypes and their long-term efficacy, HMOs may represent a promising complementary tool for childhood allergy prevention.


Subject(s)
Food Hypersensitivity , Milk, Human , Infant , Female , Humans , Child , Infant Formula/chemistry , Food Hypersensitivity/prevention & control , Breast Feeding , Oligosaccharides/therapeutic use , Oligosaccharides/analysis
16.
Nature ; 577(7791): 543-548, 2020 01.
Article in English | MEDLINE | ID: mdl-31915378

ABSTRACT

Although maternal antibodies protect newborn babies from infection1,2, little is known about how protective antibodies are induced without prior pathogen exposure. Here we show that neonatal mice that lack the capacity to produce IgG are protected from infection with the enteric pathogen enterotoxigenic Escherichia coli by maternal natural IgG antibodies against the maternal microbiota when antibodies are delivered either across the placenta or through breast milk. By challenging pups that were fostered by either maternal antibody-sufficient or antibody-deficient dams, we found that IgG derived from breast milk was crucial for protection against mucosal disease induced by enterotoxigenic E. coli. IgG also provides protection against systemic infection by E. coli. Pups used the neonatal Fc receptor to transfer IgG from milk into serum. The maternal commensal microbiota can induce antibodies that recognize antigens expressed by enterotoxigenic E. coli and other Enterobacteriaceae species. Induction of maternal antibodies against a commensal Pantoea species confers protection against enterotoxigenic E. coli in pups. This role of the microbiota in eliciting protective antibodies to a specific neonatal pathogen represents an important host defence mechanism against infection in neonates.


Subject(s)
Antibodies/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Immunity, Maternally-Acquired/immunology , Infant, Newborn/immunology , Microbiota/immunology , Milk, Human/immunology , Animals , Antibodies/blood , Antibodies/metabolism , Breast Feeding , Cross Reactions/immunology , Escherichia coli Infections/microbiology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Male , Mice , Mothers , Pantoea/immunology , Receptors, Fc/immunology , Receptors, Fc/metabolism , Symbiosis/immunology
17.
Proc Natl Acad Sci U S A ; 120(30): e2221413120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37433002

ABSTRACT

Effects of micronutrients on brain connectivity are incompletely understood. Analyzing human milk samples across global populations, we identified the carbocyclic sugar myo-inositol as a component that promotes brain development. We determined that it is most abundant in human milk during early lactation when neuronal connections rapidly form in the infant brain. Myo-inositol promoted synapse abundance in human excitatory neurons as well as cultured rat neurons and acted in a dose-dependent manner. Mechanistically, myo-inositol enhanced the ability of neurons to respond to transsynaptic interactions that induce synapses. Effects of myo-inositol in the developing brain were tested in mice, and its dietary supplementation enlarged excitatory postsynaptic sites in the maturing cortex. Utilizing an organotypic slice culture system, we additionally determined that myo-inositol is bioactive in mature brain tissue, and treatment of organotypic slices with this carbocyclic sugar increased the number and size of postsynaptic specializations and excitatory synapse density. This study advances our understanding of the impact of human milk on the infant brain and identifies myo-inositol as a breast milk component that promotes the formation of neuronal connections.


Subject(s)
Breast Feeding , Milk, Human , Female , Infant , Humans , Animals , Mice , Rats , Neurons , Inositol/pharmacology , Sugars
18.
Mol Cell Proteomics ; 22(9): 100635, 2023 09.
Article in English | MEDLINE | ID: mdl-37597722

ABSTRACT

Breast milk is abundant with functionalized milk oligosaccharides (MOs) to nourish and protect the neonate. Yet we lack a comprehensive understanding of the repertoire and evolution of MOs across Mammalia. We report ∼400 MO-species associations (>100 novel structures) from milk glycomics of nine mostly understudied species: alpaca, beluga whale, black rhinoceros, bottlenose dolphin, impala, L'Hoest's monkey, pygmy hippopotamus, domestic sheep, and striped dolphin. This revealed the hitherto unknown existence of the LacdiNAc motif (GalNAcß1-4GlcNAc) in MOs of all species except alpaca, sheep, and striped dolphin, indicating the widespread occurrence of this potentially antimicrobial motif in MOs. We also characterize glucuronic acid-containing MOs in the milk of impala, dolphins, sheep, and rhinoceros, previously only reported in cows. We demonstrate that these GlcA-MOs exhibit potent immunomodulatory effects. Our study extends the number of known MOs by >15%. Combined with >1900 curated MO-species associations, we characterize MO motif distributions, presenting an exhaustive overview of MO biodiversity.


Subject(s)
Antelopes , Camelids, New World , Dolphins , Stenella , Humans , Female , Infant, Newborn , Animals , Cattle , Sheep , Milk, Human , Oligosaccharides
19.
Proc Natl Acad Sci U S A ; 119(15): e2121720119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377806

ABSTRACT

Human breast milk (hBM) is a dynamic fluid that contains millions of cells, but their identities and phenotypic properties are poorly understood. We generated and analyzed single-cell RNA-sequencing (scRNA-seq) data to characterize the transcriptomes of cells from hBM across lactational time from 3 to 632 d postpartum in 15 donors. We found that the majority of cells in hBM are lactocytes, a specialized epithelial subset, and that cell-type frequencies shift over the course of lactation, yielding greater epithelial diversity at later points. Analysis of lactocytes reveals a continuum of cell states characterized by transcriptional changes in hormone-, growth factor-, and milk production-related pathways. Generalized additive models suggest that one subcluster, LC1 epithelial cells, increases as a function of time postpartum, daycare attendance, and the use of hormonal birth control. We identify several subclusters of macrophages in hBM that are enriched for tolerogenic functions, possibly playing a role in protecting the mammary gland during lactation. Our description of the cellular components of breast milk, their association with maternal­infant dyad metadata, and our quantification of alterations at the gene and pathway levels provide a detailed longitudinal picture of hBM cells across lactational time. This work paves the way for future investigations of how a potential division of cellular labor and differential hormone regulation might be leveraged therapeutically to support healthy lactation and potentially aid in milk production.


Subject(s)
Lactation , Milk, Human , Breast Feeding , Female , Gene Expression Profiling , Humans , Lactation/genetics , Milk, Human/cytology , Milk, Human/metabolism , RNA-Seq , Transcriptome
20.
Crit Rev Biochem Mol Biol ; 57(5-6): 562-584, 2022.
Article in English | MEDLINE | ID: mdl-36866565

ABSTRACT

Bifidobacteria are early colonizers of the human neonatal gut and provide multiple health benefits to the infant, including inhibiting the growth of enteropathogens and modulating the immune system. Certain Bifidobacterium species prevail in the gut of breastfed infants due to the ability of these microorganisms to selectively forage glycans present in human milk, specifically human milk oligosaccharides (HMOs) and N-linked glycans. Therefore, these carbohydrates serve as promising prebiotic dietary supplements to stimulate the growth of bifidobacteria in the guts of children suffering from impaired gut microbiota development. However, the rational formulation of milk glycan-based prebiotics requires a detailed understanding of how bifidobacteria metabolize these carbohydrates. Accumulating biochemical and genomic data suggest that HMO and N-glycan assimilation abilities vary remarkably within the Bifidobacterium genus, both at the species and strain levels. This review focuses on the delineation and genome-based comparative analysis of differences in respective biochemical pathways, transport systems, and associated transcriptional regulatory networks, providing a foundation for genomics-based projection of milk glycan utilization capabilities across a rapidly growing number of sequenced bifidobacterial genomes and metagenomic datasets. This analysis also highlights remaining knowledge gaps and suggests directions for future studies to optimize the formulation of milk-glycan-based prebiotics that target bifidobacteria.


Subject(s)
Bifidobacterium , Prebiotics , Infant , Infant, Newborn , Child , Humans , Bifidobacterium/genetics , Bifidobacterium/metabolism , Prebiotics/analysis , Milk, Human/chemistry , Milk, Human/metabolism , Polysaccharides/metabolism , Carbohydrates/analysis , Genomics
SELECTION OF CITATIONS
SEARCH DETAIL