Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery

Derrick M Chu, Jun Ma, Amanda L Prince, Kathleen M Antony, Maxim D Seferovic, Kjersti M Aagaard, Derrick M Chu, Jun Ma, Amanda L Prince, Kathleen M Antony, Maxim D Seferovic, Kjersti M Aagaard

Abstract

Human microbial communities are characterized by their taxonomic, metagenomic and metabolic diversity, which varies by distinct body sites and influences human physiology. However, when and how microbial communities within each body niche acquire unique taxonomical and functional signatures in early life remains underexplored. We thus sought to determine the taxonomic composition and potential metabolic function of the neonatal and early infant microbiota across multiple body sites and assess the effect of the mode of delivery and its potential confounders or modifiers. A cohort of pregnant women in their early third trimester (n = 81) were prospectively enrolled for longitudinal sampling through 6 weeks after delivery, and a second matched cross-sectional cohort (n = 81) was additionally recruited for sampling once at the time of delivery. Samples across multiple body sites, including stool, oral gingiva, nares, skin and vagina were collected for each maternal-infant dyad. Whole-genome shotgun sequencing and sequencing analysis of the gene encoding the 16S rRNA were performed to interrogate the composition and function of the neonatal and maternal microbiota. We found that the neonatal microbiota and its associated functional pathways were relatively homogeneous across all body sites at delivery, with the notable exception of the neonatal meconium. However, by 6 weeks after delivery, the infant microbiota structure and function had substantially expanded and diversified, with the body site serving as the primary determinant of the composition of the bacterial community and its functional capacity. Although minor variations in the neonatal (immediately at birth) microbiota community structure were associated with the cesarean mode of delivery in some body sites (oral gingiva, nares and skin; R2 = 0.038), this was not true for neonatal stool (meconium; Mann-Whitney P > 0.05), and there was no observable difference in community function regardless of delivery mode. For infants at 6 weeks of age, the microbiota structure and function had expanded and diversified with demonstrable body site specificity (P < 0.001, R2 = 0.189) but without discernable differences in community structure or function between infants delivered vaginally or by cesarean surgery (P = 0.057, R2 = 0.007). We conclude that within the first 6 weeks of life, the infant microbiota undergoes substantial reorganization, which is primarily driven by body site and not by mode of delivery.

Conflict of interest statement

Competing Financial Interest Statement

The authors declare no conflicts of interest.

Figures

Fig. 1. Neonatal (at birth) microbial community…
Fig. 1. Neonatal (at birth) microbial community structure
(A) Principal coordinate analysis (PCoA) on unweighted UniFrac distances between the neonatal microbiota is shown along the first two principal coordinate (PC) axes. Boxplots shown along each PC axis represents the distribution of samples along the given axis, representing the median and interquartile range with whiskers determined by Tukey’s method. Each point represents a single sample and is colored by body site: Meconium (Mec.), Orange; Skin, blue; Oral Cavity, red; Nares, green. Ellipses represent a 95% confidence interval around the cluster centroid. Clustering significance by virtue of body site was determined by Adonis (p<0.001). (B) Cumulative distribution of Bray-Curtis dissimilarity distances calculated pairwise between samples of the same body site (solid lines), and between different body sites (dashed lines). Distance comparisons for neonates and maternal samples are shown in red and gray, respectively. Smaller values indicate a greater similarity between samples. (C) The average relative abundance (circle size) of the most prevalent genera (y-axis) in each body site (x-axis) is plotted for neonates and mothers at delivery. The indicator value index (related to circle color shade darkness) represents the strength of association between a taxa and a given body site, with larger values indicating greater specificity.
Fig. 2. The infant microbiota at 6…
Fig. 2. The infant microbiota at 6 weeks demonstrates body site specificity
(A) PCoA on unweighted UniFrac distances between the infant microbiota is shown along the first two PC axes. Boxplots shown along each PC axis represents the distribution of samples along the given axis, representing the median and interquartile range with whiskers determined by Tukey’s method. Each point represents a single sample and is colored by body site: Stool, orange; Skin, blue; Oral cavity, red; Nares, green. Ellipses represent a 95% confidence interval around the cluster centroid. Clustering significance by virtue of body site was determined by Adonis (p<0.001). (B) Cumulative distribution of Bray-Curtis dissimilarity distances calculated pairwise between samples of the same body site (solid lines), and between different body sites (dashed lines). Distance comparisons for infant and maternal samples are shown in red and gray, respectively. Smaller values indicate a greater similarity between samples. (C) The average relative abundance (circle size) of the most prevalent genera (y-axis) in each body site (x-axis) is plotted for infants and mothers at 6 weeks. The indicator value index (related to circle shade darkness) represents the strength of association between a taxa and a given body site, with larger values indicating greater specificity.
Fig. 3. Failure to demonstrate a significant…
Fig. 3. Failure to demonstrate a significant impact of mode of delivery on the infant microbiota across body sites and time
(A and B) PCoA on unweighted UniFrac distances between the neonatal microbiota at delivery (A) and 6 weeks (B). Data are stratified by body site, with mode of delivery indicated by color (Cesarean, red; vaginal, gray). Ellipses represent the 95% confidence interval around the cluster centroid. Significance of clustering, which was seen among neonatal (A, delivery) nasal, oral, and skin communities, but not meconium (Mann-Whitney test of PC1 values: p>0.05), was determined by an Adonis test by virtue of mode of delivery, stratified by body site (R2=0.038). Conversely, among infants (B,6 weeks), significant clustering was not observed (Adonis p=0.057, R2=0.007). (C and D) Three axes ternary plots indicating the proportion of OTUs within a neonatal sample (each point) predicted to originate from a maternal body site (indicated by the triangle vertices). Each point represents a neonatal sample while its position indicates the predicted relative contribution from either the maternal vagina (posterior fornix or introitus), maternal skin (retroauricular crease or antecubital fossa) or from another maternal site (supragingival plaque, anterior nares, stool, unknown). Points closer to the vertices indicate that a greater proportion of the samples OTUs are predicted to originate from the microbiota of the indicated maternal body site. Data for samples obtained at delivery (C) and at 6 weeks (D) are stratified by body site and by mode of delivery (Vaginal, Cesarean-Labored, or Cesarean-Unlabored). A 2D point density topography map (blue shading) for each plot is given to indicate the point density.
Fig. 4. Taxonomic profiles of infant and…
Fig. 4. Taxonomic profiles of infant and maternal stool and oral microbiomes according to mode of delivery and time
A phylogenetic representation of the taxonomic composition in the infant stool (A), maternal stool (B), infant oral (C), and maternal oral (D) samples at delivery (inner two rings) and 6 weeks postpartum (outer two rings). Each time point is further divided by mode of delivery (indicated by the label at the bottom of the rings. The average relative abundance of each genera are plotted within the concentric rings, represented by the shaded cells, with higher relative abundance indicated by a darker shade. The phyla to which each taxa belongs is indicated by the phylogenetic tree. Three notable taxa are indicated by the red outline, namely Bacteroides, Bifidobacterium and Lactobacillus.
Fig. 5. Expansion and diversification of the…
Fig. 5. Expansion and diversification of the infant microbial community structure and function by 6 weeks of age
(A) Heatmap showing distinct microbial gene (KEGG pathway) profiles of the infant stool and oral metagenome at delivery and 6 weeks of age. The microbial gene profile for maternal stool is shown as a comparison. The relative abundance of a pathway in a given sample is colored by its row z-score ((value – row mean)/row standard deviation). The vertical color bar represents the higher order KEGG module to which each pathway belongs. (B) PCoA of Bray-Curtis distances based on pathway relative abundances demonstrates primarily clustering by body site and time point (PERMANOVA, p3.0 are shown.
Fig. 6. Infant microbial community function with…
Fig. 6. Infant microbial community function with clinical metadata in a generalized linear model
(A) Heatmap of the relative abundances of KEGG pathways found in infant stool samples as determined by WGS sequencing. The vertical bar on the left indicates mode of delivery, breastfeeding practices and intrapartum antibiotic usage. Dendrograms represent hierarchical clustering on Euclidean distances using average linkage. (B) A generalized linear mixed model was fitted for each pathway to identify pathways whose abundances differed significantly between individuals by virtue of mode of delivery, antibiotic usage, breastfeeding, gestational weight gain, BMI and gestational age. The strength of the linear model predictions for each pathway is represented by bar height. Significant correlations are indicated by the darker color (dark red or dark gray). Labels correspond to the following comparisons: Vaginal Delivery - pathways enriched in infants born vaginally (red, up) or by Cesarean (gray, down); Intrapartum Antibiotics - pathways enriched in infants exposed to intrapartum antibiotic usage (red, up) as opposed to no antibiotics (gray, down); Mixed Formula and Human Milk Feeding - pathways increased in partially breast fed (human milk and formula) infants (red, up) as opposed to exclusive human milk only (gray, down); Exclusive Formula Feeding - pathways higher in exclusively formula fed infants (red, up), as opposed to human milk only (gray, down); Excess Gestational Weight Gain - pathways higher in cases of excess maternal gestational weight gain (red, up), as opposed to normal weight gain (gray, down); Pre-pregnancy BMI & Gestational age - pathways positively (up, red) or negatively (down, gray) correlated with pre-pregnancy BMI or gestational age.

References

Main text References

    1. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214.
    1. Arumugam M, et al. Enterotypes of the human gut microbiome. Nature. 2011;473:174–180.
    1. Aagaard K, et al. A metagenomic approach to characterization of the vaginal microbiome signature in pregnancy. PloS One. 2012;7:e36466.
    1. Costello EK, et al. Bacterial community variation in human body habitats across space and time. Science. 2009;326:1694–1697.
    1. Turnbaugh PJ, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–131.
    1. Ravel J, et al. Vaginal microbiome of reproductive-age women. Proc. Natl. Acad. Sci. U. S. A. 2011;(108 Suppl 1):4680–4687.
    1. Ridaura VK, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214.
    1. Qin J, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60.
    1. Morgan XC, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79.
    1. Schulz MD, et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature. 2014;514:508–512.
    1. Gordon HA, Pesti L. The gnotobiotic animal as a tool in the study of host microbial relationships. Bacteriol. Rev. 1971;35:390–429.
    1. Olszak T, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336:489–493.
    1. Wesemann DR, et al. Microbial colonization influences early B-lineage development in the gut lamina propria. Nature. 2013;501:112–115.
    1. Gomez de Aguero M, et al. The maternal microbiota drives early postnatal innate immune development. Science. 2016;351:1296–1302.
    1. Ivanov II, et al. Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria. Cell. 2009;139:485–498.
    1. Smith PM, et al. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science. 2013;341:569–573.
    1. Atarashi K, et al. Induction of Colonic Regulatory T Cells by Indigenous Clostridium Species. Science. 2011;331:337–341.
    1. Costello EK, Carlisle EM, Bik EM, Morowitz MJ, Relman DA. Microbiome Assembly across Multiple Body Sites in Low-Birthweight Infants. mBio. 2013;4:e00782–13.
    1. Feng XL, Xu L, Guo Y, Ronsmans C. Factors influencing rising caesarean section rates in China between 1988 and 2008. Bull. World Health Organ. 2012;90:30–39.
    1. Osterman MJK, Martin JA. Trends in low-risk cesarean delivery in the United States, 1990–2013. Natl. Vital Stat. Rep. Cent. Dis. Control Prev. Natl. Cent. Health Stat. Natl. Vital Stat. Syst. 2014;63:1–16.
    1. Almqvist C, Cnattingius S, Lichtenstein P, Lundholm C. The impact of birth mode of delivery on childhood asthma and allergic diseases--a sibling study. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2012;42:1369–1376.
    1. Black M, Bhattacharya S, Philip S, Norman JE, McLernon DJ. Planned Repeat Cesarean Section at Term and Adverse Childhood Health Outcomes: A Record-Linkage Study. PLoS Med. 2016;13:e1001973.
    1. American College of Obstetricians and Gynecologists (College) et al. Safe prevention of the primary cesarean delivery. Am. J. Obstet. Gynecol. 2014;210:179–193.
    1. Dominguez-Bello MG, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. 2010;107:11971–11975.
    1. Biasucci G, et al. Mode of delivery affects the bacterial community in the newborn gut. Early Hum. Dev. 2010;(86 Suppl 1):13–15.
    1. Fallani M, et al. Intestinal microbiota of 6-week-old infants across Europe: geographic influence beyond delivery mode, breast-feeding, and antibiotics. J. Pediatr. Gastroenterol. Nutr. 2010;51:77–84.
    1. Azad MB, et al. Gut microbiota of healthy Canadian infants: profiles by mode of delivery and infant diet at 4 months. Can. Med. Assoc. J. 2013;185:385–394.
    1. Barber EL, et al. Indications Contributing to the Increasing Cesarean Delivery Rate. Obstet. Gynecol. 2011;118:29–38.
    1. David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563.
    1. Koenig JE, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. 2011;108:4578–4585.
    1. Jost T, Lacroix C, Braegger CP, Rochat F, Chassard C. Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ. Microbiol. 2014;16:2891–2904.
    1. Azad M, et al. Impact of maternal intrapartum antibiotics, method of birth and breastfeeding on gut microbiota during the first year of life: a prospective cohort study. BJOG Int. J. Obstet. Gynaecol. 2015 n/a-n/a.
    1. Ardeshir A, et al. Breast-fed and bottle-fed infant rhesus macaques develop distinct gut microbiotas and immune systems. Sci. Transl. Med. 2014;6:252ra120.
    1. Dufrene M, Legendre P. Species Assemblages and Indicator Species: The Need for a Flexible Asymmetrical Approach. Ecological Monographs. 1997;67:345–366.
    1. Koren O, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell. 2012;150:470–480.
    1. La Rosa PS, et al. Patterned progression of bacterial populations in the premature infant gut. Proc. Natl. Acad. Sci. U. S. A. 2014;111:12522–12527.
    1. Aagaard K, et al. The placenta harbors a unique microbiome. Sci. Transl. Med. 2014;6:237ra65.
    1. Antony KM, et al. The preterm placental microbiome varies in association with excess maternal gestational weight gain. Am. J. Obstet. Gynecol. 2015;212:653.e1–16.
    1. Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci. Rep. 2016;6:23129.
    1. Bäckhed F, et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe. 2015;17:852.
    1. Knights D, et al. Bayesian community-wide culture-independent microbial source tracking. Nat. Methods. 2011;8:761–763.
    1. Penders J, et al. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics. 2006;118:511–521.
    1. Vatanen T, et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell. 2016;165:842–853.
    1. Dominguez-Bello MG, et al. Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat. Med. 2016;22:250–253.
    1. Galley JD, Bailey M, Kamp Dush C, Schoppe-Sullivan S, Christian LM. Maternal obesity is associated with alterations in the gut microbiome in toddlers. PloS One. 2014;9:e113026.
    1. Mueller NT, et al. Birth mode-dependent association between pre-pregnancy maternal weight status and the neonatal intestinal microbiome. Sci. Rep. 2016;6:23133.
    1. Chu DM, et al. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med. 2016;8:77.
    1. Hu J, et al. Diversified Microbiota of Meconium Is Affected by Maternal Diabetes Status. PLoS One. 2013;8:e78257.
    1. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr. Opin. Gastroenterol. 2014;30:332–338.
    1. Kennedy D. B Vitamins and the Brain: Mechanisms, Dose and Efficacy—A Review. Nutrients. 2016;8:68.
    1. Gibson MK, et al. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat. Microbiol. 2016;1:16024.
    1. Bokulich NA, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016;8:343ra82–343ra82.
    1. DiGiulio DB, et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PloS One. 2008;3:e3056.
    1. Steel JH, et al. Bacteria and inflammatory cells in fetal membranes do not always cause preterm labor. Pediatr. Res. 2005;57:404–411.
    1. Jiménez E, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr. Microbiol. 2005;51:270–274.
    1. Jiménez E, et al. Is meconium from healthy newborns actually sterile? Res. Microbiol. 2008;159:187–193.
    1. Ardissone AN, et al. Meconium microbiome analysis identifies bacteria correlated with premature birth. PloS One. 2014;9:e90784.
    1. Hansen R, et al. First-Pass Meconium Samples from Healthy Term Vaginally-Delivered Neonates: An Analysis of the Microbiota. PLOS One. 2015;10:e0133320.
    1. Fallani M, et al. Determinants of the human infant intestinal microbiota after the introduction of first complementary foods in infant samples from five European centres. Microbiology. 2011;157:1385–1392.
    1. Yassour M, et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 2016;8:343ra81–343ra81.
Methods-only References
    1. La Rosa PS, et al. Hypothesis Testing and Power Calculations for Taxonomic-Based Human Microbiome Data. PLoS One. 2012;7:e52078.
    1. Aagaard K, et al. The Human Microbiome Project strategy for comprehensive sampling of the human microbiome and why it matters. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2013;27:1012–1022.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–336.
    1. Schmieder R, Edwards R. Fast identification and removal of sequence contamination from genomic and metagenomic datasets. PloS One. 2011;6:e17288.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinforma. Oxf. Engl. 2010;26:2460–2461.
    1. Haas BJ, et al. Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res. 2011;21:494–504.
    1. Cole JR, et al. The Ribosomal Database Project: improved alignments and new tools for rRNA analysis. Nucleic Acids Res. 2009;37:D141–D145.
    1. DeSantis TZ, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol. 2006;72:5069–5072.
    1. Segata N, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60.
    1. Segata N, et al. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat. Methods. 2012;9:811–814.
    1. Abubucker S, et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol. 2012;8:e1002358.
    1. Le Chatelier E, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–546.

Source: PubMed

3
Subscribe