Longitudinal analysis of the premature infant intestinal microbiome prior to necrotizing enterocolitis: a case-control study

Yanjiao Zhou, Gururaj Shan, Erica Sodergren, George Weinstock, W Allan Walker, Katherine E Gregory, Yanjiao Zhou, Gururaj Shan, Erica Sodergren, George Weinstock, W Allan Walker, Katherine E Gregory

Abstract

Necrotizing enterocolitis (NEC) is an inflammatory disease of the newborn bowel, primarily affecting premature infants. Early intestinal colonization has been implicated in the pathogenesis of NEC. The objective of this prospective case-control study was to evaluate differences in the intestinal microbiota between infants who developed NEC and unaffected controls prior to disease onset. We conducted longitudinal analysis of the 16S rRNA genes of 312 samples obtained from 12 NEC cases and 26 age-matched controls with a median frequency of 7 samples per subject and median sampling interval of 3 days. We found that the microbiome undergoes dynamic development during the first two months of life with day of life being the major factor contributing to the colonization process. Depending on when the infant was diagnosed with NEC (i.e. early vs. late onset), the pattern of microbial progression was different for cases and controls. The difference in the microbiota was most overt in early onset NEC cases and controls. In proximity to NEC onset, the abundances of Clostridium sensu stricto from Clostridia class were significantly higher in early onset NEC subjects comparing to controls. In late onset NEC, Escherichia/Shigella among Gammaproteobacteria, showed an increasing pattern prior to disease onset, and was significantly higher in cases than controls six days before NEC onset. Cronobacter from Gammaproteobacteria was also significantly higher in late onset NEC cases than controls 1-3 days prior to NEC onset. Thus, the specific infectious agent associated with NEC may vary by the age of infant at disease onset. We found that intravenously administered antibiotics may have an impact on the microbial diversity present in fecal material. Longitudinal analysis at multiple time points was an important strategy utilized in this study, allowing us to appreciate the dynamics of the premature infant intestinal microbiome while approaching NEC at various points.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Premature gut microbiota variation in…
Fig 1. Premature gut microbiota variation in the first two month of life.
The gut microbiota variation at class level over 0–60 days of life from all the samples is visualized by NMDS plot. The color gradient from red to blue represents the day of life of the babies. The microbiota from early day of life is distinguished from the elder days.
Fig 2. NMDS of early and late…
Fig 2. NMDS of early and late onset NEC and controls at the genus level.
The difference between NEC and controls is displayed for early onset NEC (Fig. 2A) and late onset NEC (Fig. 2B) with their controls by NMDS plot. Each dot represents one sample. Green dots represent the controls and red dots represent the NEC samples. Early NEC subjects and control subjects have a clear separation at second week of the life. The distinction between late onset NEC and controls is less obvious except at the third week of life.
Fig 3. Microbiota progression before early onset…
Fig 3. Microbiota progression before early onset NEC and late onset NEC at class and genus level.
The relative abundances of four most dominant classes (Fig. 3A-early onset at class level, 3B-late onset at class level) and the genera (Fig. 3C-early onset at genus level, 3D-late onset at genus level) that are significantly different between NEC and controls are plotted at 7–9 days, 4–6 days and 1–3 days prior to NEC onset. In early onset NEC category, 3,6, 8 NEC samples were included at 7–9 days, 4–6 days and 1–3 days; 8, 5, 6 control samples were included in the above time points. In late onset NEC category, 7, 7,10 NEC samples and 6, 8, 4 control samples were included in the above time points. Red and green of the boxplots indicate NEC and control samples, respectively. The asterisks indicate the significant difference between NEC and control.

References

    1. Costello EK, Carlisle EM, Bik EM, Morowitz MJ, Relman DA. Microbiome assembly across multiple body sites in low-birthweight infants. MBio. 2013; 4: e00782–00713. 10.1128/mBio.00782-13
    1. Madan JC, Salari RC, Saxena D, Davidson L, O’Toole GA, Moore JH, et al. Gut microbial colonisation in premature neonates predicts neonatal sepsis. Arch Dis Child Fetal Neonatal Ed. 2012; 97: F456–462. 10.1136/fetalneonatal-2011-301373
    1. Morowitz MJ, Denef VJ, Costello EK, Thomas BC, Poroyko V, Relman DA, et al. Strain-resolved community genomic analysis of gut microbial colonization in a premature infant. Proc Natl Acad Sci U S A. 2011; 108: 1128–1133. 10.1073/pnas.1010992108
    1. Sharon I, Morowitz MJ, Thomas BC, Costello EK, Relman DA, Banfield JF, et al. Time series community genomics analysis reveals rapid shifts in bacterial species, strains, and phage during infant gut colonization. Genome Res. 2013; 23: 111–120. 10.1101/gr.142315.112
    1. Nanthakumar NN, Fusunyan RD, Sanderson I, Walker WA. Inflammation in the developing human intestine: A possible pathophysiologic contribution to necrotizing enterocolitis. Proc Natl Acad Sci U S A. 2000; 97: 6043–6048.
    1. Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med. 2011; 364: 255–264. 10.1056/NEJMra1005408
    1. Wadhawan R, Oh W, Hintz SR, Blakely ML, Das A, Bell EF, et al. Neurodevelopmental outcomes of extremely low birth weight infants with spontaneous intestinal perforation or surgical necrotizing enterocolitis. J Perinatol. 2014; 34: 64–70. 10.1038/jp.2013.128
    1. Holman RC, Stoll BJ, Curns AT, Yorita KL, Steiner CA, Schonberger LB, et al. Necrotising enterocolitis hospitalisations among neonates in the United States. Paediatr Perinat Epidemiol. 2006; 20: 498–506.
    1. Stoll BJ, Hansen NI, Bell EF, Shankaran S, Laptook AR, Walsh MC, et al. Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics. 2010; 126: 443–456. 10.1542/peds.2009-2959
    1. Normann E, Fahlen A, Engstrand L, Lilja HE. Intestinal microbial profiles in extremely preterm infants with and without necrotizing enterocolitis. Acta Paediatr. 2013; 102: 129–136. 10.1111/apa.12059
    1. Mshvildadze M, Neu J, Shuster J, Theriaque D, Li N, Mai V, et al. Intestinal microbial ecology in premature infants assessed with non-culture-based techniques. J Pediatr. 2010; 156: 20–25. 10.1016/j.jpeds.2009.06.063
    1. Claud EC, Keegan KP, Brulc JM, Lu L, Bartels D, Glass E, et al. Bacterial community structure and functional contributions to emergence of health or necrotizing enterocolitis in preterm infants. Microbiome. 2013; 1: 20 10.1186/2049-2618-1-20
    1. Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, et al. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. ISME. 2009; 3: 944–954. 10.1038/ismej.2009.37
    1. Torrazza RM, Ukhanova M, Wang X, Sharma R, Hudak ML, Neu J, et al. Intestinal microbial ecology and environmental factors affecting necrotizing enterocolitis. PLoS One. 2013; 8: e83304 10.1371/journal.pone.0083304
    1. Morrow AL, Lagomarcino AJ, Schibler KR, Taft DH, Yu Z, Wang B, et al. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. Microbiome. 2013; 1: 13 10.1186/2049-2618-1-13
    1. Mai V, Young CM, Ukhanova M, Wang X, Sun Y, Casella G, et al. Fecal microbiota in premature infants prior to necrotizing enterocolitis. PLoS One. 2011; 6: e20647 10.1371/journal.pone.0020647
    1. Walsh MC, Kliegman RM. Necrotizing enterocolitis: treatment based on staging criteria. Pediatr Clin North Am. 1986; 33: 179–201.
    1. Gordon PV, Swanson JR, Attridge JT, Clark R. Emerging trends in acquired neonatal intestinal disease: is it time to abandon Bell’s criteria? J Perinatol. 2007; 27: 661–671.
    1. The Human Microbiome Consortium. A framework for human microbiome research. Nature. 2012; 486: 215–221. 10.1038/nature11209
    1. Haas BJ, Gevers D, Earl AM, Feldgarden M, Ward DV, Giannoukos G, et al. Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res. 2011; 21: 494–504. 10.1101/gr.112730.110
    1. Oksanen J, Blanchet FG, Kindt R, Legendre P, Minchin PR, O’Hara RB, et al. Vegan: Community Ecology Package. R package version 2.0–7. Available: (2013).
    1. White JR, Nagarajan N, Pop M. Statistical methods for detection differentially abundant features in clinical metagenomic samples. PLoS Computational Biology. 2009; 5: e1000352 10.1371/journal.pcbi.1000352
    1. Zhou Y, Gao H, Mihindukulasuriya KA, La Rosa PS, Wylie KM, Vishnivetskaya T, et al. Biogeography of the ecosystems of the healthy human body. Genome Biol. 2013; 14: R1 10.1186/gb-2013-14-1-r1
    1. Bates D, Maechler M, Bolker B. lme4: Linear mixed-effects models using S4 classes. R package version 0.999999–0. Available: (2012)
    1. Collins MD, Lawson PA, Willems A, Cordoba JJ, Fernandez-Garayzabal J, Garcia P, et al. (The phylogeny of the genus Clostridium: proposal of five new genera and eleven new species combinations. Int J Syst Bacteriol. 1994; 44: 812–826.
    1. Perez-Cobas AE, Gosalbes MJ, Friedrichs A, Knecht H, Artacho A, Eismann K, et al. Gut microbiota disturbance during antibiotic therapy: a multi-omic approach. Gut. 2013; 62: 1591–1601. 10.1136/gutjnl-2012-303184
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012; 486: 222–227. 10.1038/nature11053
    1. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human infant intestinal microbiota. PLoS Biol. 2007; 5: e177
    1. LaRosa P, Warner B, Zhou Y, Weinstock G, Sodergren E, Hall-Moore CM, et al. The Patterned Progression of Bacterial Populations in the Premature Infant Gut. Proc Natl Acad Sci U S A. 2014; 111: 12522–12527. 10.1073/pnas.1409497111
    1. Grishin A, Papillon S, Bell B, Wang J, Ford HR. The role of the intestinal microbiota in the pathogenesis of necrotizing enterocolitis. Semin Pediatr Surg. 2013; 22: 69–75. 10.1053/j.sempedsurg.2013.01.002
    1. Hunter CJ, Singamsetty VK, Chokshi NK, Boyle P, Camerini V, Grishin AV, et al. Enterobacter sakazakii enhances epithelial cell injury by inducing apoptosis in a rat model of necrotizing enterocolitis. J Infect Dis. 2008; 198: 586–593. 10.1086/590186
    1. Emami CN, Mittal R, Wang L, Ford HR, Prasadarao NV. Role of neutrophils and macrophages in the pathogenesis of necrotizing enterocolitis caused by Cronobacter sakazakii. J Surg Res. 2012; 172: 18–28. 10.1016/j.jss.2011.04.019
    1. Weng M, Ganguli K, Zhu W, Shi HN, Walker WA. Conditioned medium from Bifidobacteria infantis protects against Cronobacter sakazakii-induced intestinal inflammation in newborn mice. Am J Physiol Gastrointest Liver Physiol. 2014; 306: G779–787. 10.1152/ajpgi.00183.2013
    1. Jenke AC, Postberg J, Mariel B, Hensel K, Foell D, Däbritz J, et al. S100A12 and hBD2 correlate with the composition of the fecal microflora in ELBW infants and expansion of E. coli is associated with NEC. Biomed Res Int. 2013; 150372 10.1155/2013/150372
    1. Mai V, Torrazza RM, Ukhanova M, Wang X, Sun Y, Li N, et al. Distortions in development of intestinal microbiota associated with late onset sepsis in preterm infants. PLoS One. 2013; 8: e52876 10.1371/journal.pone.0052876
    1. Hallstrom M, Eerola E, Vuento R, Janas M, Tammela O. Effects of mode of delivery and necrotising enterocolitis on the intestinal microflora in preterm infants. Eur J Clin Microbiol Infect Dis. 2004; 23: 463–470.
    1. de la Cochetiere MF, Piloquet H, des Robert C, Darmaun D, Galmiche JP, Roze JC, et al. (Early intestinal bacterial colonization and necrotizing enterocolitis in premature infants: the putative role of Clostridium. Pediatr Res. 2004; 56: 366–370.
    1. Elahi S, Ertelt JM, Kinder JM, Jiang TT, Zhang X, Xin L, et al. Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection. Nature. 2013; 504: 158–162. 10.1038/nature12675
    1. Goodrich JK, Di Rienzi SC, Poole AC, Koren O, Walters WA, Caporaso JG, et al. Conducting a microbiome study. Cell. 2014; 158: 250–262. 10.1016/j.cell.2014.06.037
    1. Sullivan S, Schanler RJ, Kim JH, Patel AL, Trawoger R, Kiechl-Kohlendorfer U, et al. An exclusively human milk-based diet is associated with a lower rate of necrotizing enterocolitis than a diet of human milk and bovine milk-based products. J Pediatr. 2010; 156: 562–567.e561. 10.1016/j.jpeds.2009.10.040
    1. Ardeshir A, Narayan NR, Méndez-Lagares G, Lu D, Rauch M, Huang Y, et al. Breast-fed and bottle-fed infant rhesus macaques develop distinct gut microbiotas and immune systems. Science Translational Medicine. 2014; 6: 252ra120 10.1126/scitranslmed.3008791
    1. O’Sullivan A, He X, McNiven EM, Haggarty NW, Lonnerdal B, Slupsky CM, et al. Early diet impacts infant rhesus gut microbiome, immunity, and metabolism. J Proteome Res. 2013; 12: 2833–2845. 10.1021/pr4001702

Source: PubMed

3
Abonnieren