Microbes in Infant Gut Development: Placing Abundance Within Environmental, Clinical and Growth Parameters

Tanja Obermajer, Iztok Grabnar, Evgen Benedik, Tina Tušar, Tatjana Robič Pikel, Nataša Fidler Mis, Bojana Bogovič Matijašić, Irena Rogelj, Tanja Obermajer, Iztok Grabnar, Evgen Benedik, Tina Tušar, Tatjana Robič Pikel, Nataša Fidler Mis, Bojana Bogovič Matijašić, Irena Rogelj

Abstract

Sound and timely microbial gut colonization completes newborn's healthy metabolic programming and manifests in infant appropriate growth and weight development. Feces, collected at 3, 30, and 90 days after birth from 60 breastfed Slovenian newborns, was submitted to microbial DNA extraction and qPCR quantification of selected gut associated taxa. Multivariate regression analysis was applied to evaluate microbial dynamics with respect to infant demographic, environmental, clinical characteristics and first year growth data. Early microbial variability was marked by the proportion of Bacilli, but diminished and converged in later samples, as bifidobacteria started to prevail. The first month proportions of enterococci were associated with maternity hospital locality and supplementation of breastfeeding with formulae, while Enterococcus faecalis proportion reflected the mode of delivery. Group Bacteroides-Prevotella proportion was associated with infant weight and ponderal index at first month. Infant mixed feeding pattern and health issues within the first month revealed the most profound and extended microbial perturbations. Our findings raise concerns over the ability of the early feeding supplementation to emulate and support the gut microbiota in a way similar to the exclusively breastfed infants. Additionally, practicing supplementation beyond the first month also manifested in higher first year weight and weight gain Z-score.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
(a,b) Relative quantification of bacteria by real-time PCR in the feces of newborns in the first 3 months. The box and whiskers plots represent the medians and interquartile ranges; error bars 10th and 90th percentiles, filled circles outliers. Asterisks denote significant differences between time-points (*p < 0.05; **p < 0.01). The results are presented as a number of group specific bacterial DNA copies in g of feces (a), as ratio group specific DNA/all bacterial DNA (%) (b).
Figure 2
Figure 2
(a,b) Principal component analysis (PCA) with varimax rotation of the fecal microbiota in infants. Loadings of relative bacterial abundances (a). This figure shows the two primary principal components, which explain 32.8% and 16.2% of data variation. Individual PCA scores at 3, 30, and 90 days after birth (b).
Figure 3
Figure 3
Diagram presenting values for the exposed significant associations between the infant growth parameters and presumptive perinatal growth confounders.

References

    1. Clarke G, O’Mahony S, Dinan T, Cryan J. Priming for health: gut microbiota acquired in early life regulates physiology, brain and behaviour. Acta Paediatr. 2014;103:812–819. doi: 10.1111/apa.12674.
    1. Cox LM, et al. Altering the Intestinal Microbiota during a Critical Developmental Window Has Lasting Metabolic Consequences. Cell. 2014;158:705–721. doi: 10.1016/j.cell.2014.05.052.
    1. Houghteling PD, Walker WA. Why is initial bacterial colonization of the intestine important to infants’ and children’s health? J. Pediatr. Gastroenterol. Nutr. 2015;60:294–307. doi: 10.1097/MPG.0000000000000597.
    1. Koleva PT, Bridgman SL, Kozyrskyj AL. The Infant Gut Microbiome: Evidence for Obesity Risk and Dietary Intervention. Nutrients. 2015;7:2237–2260. doi: 10.3390/nu7042237.
    1. Barker DJP. Developmental origins of chronic disease. Public Health. 2012;126:185–189. doi: 10.1016/j.puhe.2011.11.014.
    1. Bouret S, Levin BE, Ozanne SE. Gene-environment interactions controlling energy and glucose homeostasis and the developmental origins of obesity. Physiol. Rev. 2015;95:47–82. doi: 10.1152/physrev.00007.2014.
    1. Gohir W, Ratcliffe EM, Sloboda DM. Of the bugs that shape us: maternal obesity, the gut microbiome, and long-term disease risk. Pediatr. Res. 2015;77:196–204. doi: 10.1038/pr.2014.169.
    1. Salgin B, et al. Even transient rapid infancy weight gain is associated with higher BMI in young adults and earlier menarche. Int. J. Obes. 2015;39:939–944. doi: 10.1038/ijo.2015.25.
    1. Borre YE, et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol. Med. 2014;20:509–518. doi: 10.1016/j.molmed.2014.05.002.
    1. Nylund L, Satokari R, Salminen S, de Vos WM. Intestinal microbiota during early life - impact on health and disease. Proc. Nutr. Soc. 2014;73:457–469. doi: 10.1017/S0029665114000627.
    1. Dogra S, et al. Rate of establishing the gut microbiota in infancy has consequences for future health. Gut Microbes. 2015;6:321–325. doi: 10.1080/19490976.2015.1078051.
    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. doi: 10.1097/MPG.0b013e3181d1b11e.
    1. van Best N, Hornef MW, Savelkoul PHM, Penders J. On the origin of species: Factors shaping the establishment of infant’s gut microbiota. Birth Defects Res. Part C Embryo Today Rev. 2015;105:240–251. doi: 10.1002/bdrc.21113.
    1. Wang X, et al. Multivariate Approach for Studying Interactions between Environmental Variables and Microbial Communities. PLoS ONE. 2012;7:e50267. doi: 10.1371/journal.pone.0050267.
    1. Gerber GK. The dynamic microbiome. FEBS Lett. 2014;588:4131–4139. doi: 10.1016/j.febslet.2014.02.037.
    1. Rodríguez, J. M. et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 26 (2015).
    1. Obermajer T, et al. Colostrum of Healthy Slovenian Mothers: Microbiota Composition and Bacteriocin Gene Prevalence. PLOS ONE. 2015;10:e0123324. doi: 10.1371/journal.pone.0123324.
    1. Adlerberth I, et al. Reduced Enterobacterial and Increased Staphylococcal Colonization of the Infantile Bowel: An Effect of Hygienic Lifestyle? Pediatr. Res. 2006;59:96–101. doi: 10.1203/01.pdr.0000191137.12774.b2.
    1. Arboleya S, et al. Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol. Ecol. 2012;79:763–772. doi: 10.1111/j.1574-6941.2011.01261.x.
    1. Penders J, et al. Factors Influencing the Composition of the Intestinal Microbiota in Early Infancy. Pediatrics. 2006;118:511–521. doi: 10.1542/peds.2005-2824.
    1. Dogra S, et al. Dynamics of Infant Gut Microbiota Are Influenced by Delivery Mode and Gestational Duration and Are Associated with Subsequent Adiposity. mBio. 2015;6:e02419–14. doi: 10.1128/mBio.02419-14.
    1. Rosa PSL, et al. Patterned progression of bacterial populations in the premature infant gut. Proc. Natl. Acad. Sci. 2014;111:12522–12527. doi: 10.1073/pnas.1409497111.
    1. Savage T, et al. Increasing Maternal Age Is Associated with Taller Stature and Reduced Abdominal Fat in Their Children. PLoS ONE. 2013;8:e58869. doi: 10.1371/journal.pone.0058869.
    1. Lubetzky R, Sever O, Mimouni FB, Mandel D. Human Milk Macronutrients Content: Effect of Advanced Maternal Age. Breastfeed. Med. 2015;10:433–436. doi: 10.1089/bfm.2015.0072.
    1. Hunt KM, et al. Human Milk Oligosaccharides Promote the Growth of Staphylococci. Appl. Environ. Microbiol. 2012;78:4763–4770. doi: 10.1128/AEM.00477-12.
    1. Alderete, T. L. et al. Associations between human milk oligosaccharides and infant body composition in the first 6 mo of life. Am. J. Clin. Nutr. ajcn115451 (2015).
    1. Al Atya AK, et al. Probiotic potential of Enterococcus faecalis strains isolated from meconium. Front. Microbiol. 2015;6:227. doi: 10.3389/fmicb.2015.00227.
    1. Wang S, Hibberd ML, Pettersson S, Lee YK. Enterococcus faecalis from Healthy Infants Modulates Inflammation through MAPK Signaling Pathways. PLoS ONE. 2014;9:e97523. doi: 10.1371/journal.pone.0097523.
    1. Jakobsson HE, et al. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut. 2014;63:559–566. doi: 10.1136/gutjnl-2012-303249.
    1. Kuhle S, Tong OS, Woolcott CG. Association between caesarean section and childhood obesity: a systematic review and meta-analysis. Obes. Rev. 2015;16:295–303. doi: 10.1111/obr.12267.
    1. Riva A, et al. Pediatric obesity is associated with an altered gut microbiota and discordant shifts in Firmicutes populations. Environ. Microbiol. 2017;19:95–105. doi: 10.1111/1462-2920.13463.
    1. White RA, et al. Novel Developmental Analyses Identify Longitudinal Patterns of Early Gut Microbiota that Affect Infant Growth. PLoS Comput Biol. 2013;9:e1003042. doi: 10.1371/journal.pcbi.1003042.
    1. Scheepers LEJM, et al. The intestinal microbiota composition and weight development in children: the KOALA Birth Cohort Study. Int. J. Obes. 2015;39:16–25. doi: 10.1038/ijo.2014.178.
    1. Kozyrskyj AL, Kalu R, Koleva PT, Bridgman SL. Fetal programming of overweight through the microbiome: boys are disproportionately affected. J. Dev. Orig. Health Dis. 2016;7:25–34. doi: 10.1017/S2040174415001269.
    1. Wang M, et al. Fecal microbiota composition of breast-fed infants is correlated with human milk oligosaccharides consumed. J. Pediatr. Gastroenterol. Nutr. 2015;60:825–833. doi: 10.1097/MPG.0000000000000752.
    1. O’Sullivan A, Farver M, Smilowitz JT. The Influence of Early Infant-Feeding Practices on the Intestinal Microbiome and Body Composition in Infants. Nutr. Metab. Insights. 2015;8:1–9.
    1. Charbonneau MR, et al. Sialylated Milk Oligosaccharides Promote Microbiota-Dependent Growth in Models of Infant Undernutrition. Cell. 2016;164:859–871. doi: 10.1016/j.cell.2016.01.024.
    1. Kramer MS, et al. Feeding effects on growth during infancy∗. J. Pediatr. 2004;145:600–605. doi: 10.1016/j.jpeds.2004.06.069.
    1. Bäckhed F, et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe. 2015;17:690–703. doi: 10.1016/j.chom.2015.04.004.
    1. Korpela K, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat. Commun. 2016;7:10410. doi: 10.1038/ncomms10410.
    1. Trasande L, et al. Infant antibiotic exposures and early-life body mass. Int. J. Obes. 2013;37:16–23. doi: 10.1038/ijo.2012.132.
    1. Sinkiewicz G, Ljunggren L. Occurrence of Lactobacillus reuteri in human breast milk. Microb. Ecol. Health Dis. 2008;20:122–126. doi: 10.1080/08910600802341007.
    1. Taghizadeh M, et al. The influence of impact delivery mode, lactation time, infant gender, maternal age and rural or urban life on total number of Lactobacillus in breast milk Isfahan - Iran. Adv. Biomed. Res. 2015;4:141. doi: 10.4103/2277-9175.161546.
    1. Soto A, et al. Lactobacilli and bifidobacteria in human breast milk: influence of antibiotherapy and other host and clinical factors. J. Pediatr. Gastroenterol. Nutr. 2014;59:78–88. doi: 10.1097/MPG.0000000000000347.
    1. Biedermann L, et al. Smoking Cessation Induces Profound Changes in the Composition of the Intestinal Microbiota in Humans. PLoS ONE. 2013;8:e59260. doi: 10.1371/journal.pone.0059260.
    1. Rodríguez JM. The Origin of Human Milk Bacteria: Is There a Bacterial Entero-Mammary Pathway during Late Pregnancy and Lactation? Adv. Nutr. Int. Rev. J. 2014;5:779–784. doi: 10.3945/an.114.007229.
    1. Agostoni C, et al. Earlier smoking habits are associated with higher serum lipids and lower milk fat and polyunsaturated fatty acid content in the first 6 months of lactation. Eur. J. Clin. Nutr. 2003;57:1466–1472. doi: 10.1038/sj.ejcn.1601711.
    1. Munblit D, et al. Exposures influencing total IgA level in colostrum. J. Dev. Orig. Health Dis. 2016;7:61–67. doi: 10.1017/S2040174415001476.
    1. Slaughter MH, et al. Skinfold equations for estimation of body fatness in children and youth. Hum. Biol. 1988;60:709–723.
    1. WHO Multicentre Growth Reference Study Group. WHO Child Growth Standards: Length/height-for-age, weight-for-age, weight-for-length, weight-for-height and body mass index-for-age: Methods and development. Geneva: World Health Organization. WHO Available at: . (Accessed: 5th April 2016) (2006).
    1. Pan, H. & Cole, T. J. LMSgrowth, a Microsoft Excel add-in to access growth references based on the LMS method. Version 2.77. (2012).
    1. Monteiro POA, Victora CG. Rapid growth in infancy and childhood and obesity in later life – a systematic review. Obes. Rev. 2005;6:143–154. doi: 10.1111/j.1467-789X.2005.00183.x.
    1. Matijašić BB, et al. Association of dietary type with fecal microbiota in vegetarians and omnivores in Slovenia. Eur. J. Nutr. 2013;53:1051–1064.
    1. Bartosch S, Fite A, Macfarlane GT, McMurdo MET. Characterization of Bacterial Communities in Feces from Healthy Elderly Volunteers and Hospitalized Elderly Patients by Using Real-Time PCR and Effects of Antibiotic Treatment on the Fecal Microbiota. Appl. Environ. Microbiol. 2004;70:3575–3581. doi: 10.1128/AEM.70.6.3575-3581.2004.
    1. Song Y-L, et al. Rapid identification of 11 human intestinal Lactobacillus species by multiplex PCR assays using group- and species-specific primers derived from the 16S–23S rRNA intergenic spacer region and its flanking 23S rRNA. FEMS Microbiol. Lett. 2000;187:167–173.

Source: PubMed

3
Abonnieren