Antibiotics and the developing intestinal microbiome, metabolome and inflammatory environment in a randomized trial of preterm infants

Jordan T Russell, J Lauren Ruoss, Diomel de la Cruz, Nan Li, Catalina Bazacliu, Laura Patton, Kelley Lobean McKinley, Timothy J Garrett, Richard A Polin, Eric W Triplett, Josef Neu, Jordan T Russell, J Lauren Ruoss, Diomel de la Cruz, Nan Li, Catalina Bazacliu, Laura Patton, Kelley Lobean McKinley, Timothy J Garrett, Richard A Polin, Eric W Triplett, Josef Neu

Abstract

Antibiotic use in neonates can have detrimental effects on the developing gut microbiome, increasing the risk of morbidity. A majority of preterm neonates receive antibiotics after birth without clear evidence to guide this practice. Here microbiome, metabolomic, and immune marker results from the routine early antibiotic use in symptomatic preterm Neonates (REASON) study are presented. The REASON study is the first trial to randomize symptomatic preterm neonates to receive or not receive antibiotics in the first 48 h after birth. Using 16S rRNA sequencing of stool samples collected longitudinally for 91 neonates, the effect of such antibiotic use on microbiome diversity is assessed. The results illustrate that type of nutrition shapes the early infant gut microbiome. By integrating data for the gut microbiome, stool metabolites, stool immune markers, and inferred metabolic pathways, an association was discovered between Veillonella and the neurotransmitter gamma-aminobutyric acid (GABA). These results suggest early antibiotic use may impact the gut-brain axis with the potential for consequences in early life development, a finding that needs to be validated in a larger cohort.Trial Registration This project is registered at clinicaltrials.gov under the name "Antibiotic 'Dysbiosis' in Preterm Infants" with trial number NCT02784821.

Conflict of interest statement

Dr. Josef Neu is the principal investigator of a study with Infant Bacterial Therapeutics and on the Scientific Advisory Boards of Medela and Astarte. No other authors have competing interest to disclose.

Figures

Figure 1
Figure 1
Antibiotic use 48 h after birth does not significantly affect alpha diversity development. Boxplots displaying (A) the observed ASV richness (B) the Shannon diversity and (C) log10-transformed copies of 16S rRNA by enrollment group across corrected gestational ages between weeks 28 and 39. P values were calculated at each corrected GA timepoint between enrollment groups using the non-parametric Kruskal–Wallis test. Linear mixed-effects modeling of the (D) observed ASV richness and (E) Shannon diversity over time between enrollment groups. Time scale on the x-axis is days of life (DOL) for corrected GA weeks 28–39. Greyed areas around each regression line represent 95% confidence intervals upper and lower around the coefficients.
Figure 2
Figure 2
Antibiotic use explains little effect on beta diversity. PCoA ordination of stool samples using the (A) abundance-based Bray–Curtis and (B) presence/absence-based Jaccard distance metric among enrollment groups of corrected GA between 28–39. Ellipses are calculated based on a 95% confidence interval of a multivariate t-distribution.
Figure 3
Figure 3
Effects of feeding patterns on the gut microbiome are transient over time. Effect of various feeding patterns on the (A) observed ASV richness (B) Shannon diversity and (C) log10-transformed copies of 16S rRNA by enrollment group over corrected GA from weeks 28–39. Only feeding patterns that have at least 2 samples at each timepoint were kept. Statistical comparison of feeding patterns at each corrected GA timepoint was performed using the non-parametric Kruskal–Wallis test.
Figure 4
Figure 4
Linear mixed effects modelling identifies feeding effects by group over time. Linear mixed-effects results plotted as observed bacterial richness over time by group and feeding types (A–F) and the Shannon diversity over time by group and feeding types (G–L). The number of samples used in this analysis by group and by feeding type are listed in Supplementary Table S3.
Figure 5
Figure 5
Integration of clinical and laboratory data gives detailed view of infant stay in NICU. Extensive clinical and laboratory data, when combined, provide a detailed summary of each infant’s stay in the NICU. Data included in each chart from top to bottom include: the infant ID, group assignment, antibiotic change status (bail), gestational age, any adverse clinical events (which are further described in Supplementary Figure S1), the type and duration of antibiotic use (if any), the copy-number corrected absolute composition of each weekly stool sample and it’s log10-scale number of bacterial 16S rRNA copies, the type and duration of each feeding including administration of human milk fortifier, the relative levels of C-reactive protein measured from blood, and relative concentrations of measured stool immune markers (for infants where these measurements were performed). DBM: donor breast milk, MBM: mother’s breast milk, NPO: no enteral nutrition, CRP: C-reactive protein, EGF: epidermal growth factor. Listed below is a key for the color-coded bacterial taxa used in the stool 16S rRNA copy-number corrected composition pie chart for each infant chart. A key for the bacterial color codes, adverse clinical events (including infections by body site) and the administration of human milk fortifier for each infant chart is given below.
Figure 6
Figure 6
Metabolites in stool correlate with abundance of bacterial genera. (A) Heatmap of repeated measures correlation coefficients between peak response heights of identified metabolites in stool and the top 10 bacterial genera from the same samples (n = 90 stool samples). Significant correlations are indicated by a ‘ + ’ with FDR-corrected p values < 0.05. (B) Boxplot comparing the peak response heights for 4-aminobutyric acid (GABA) between enrollment groups. Statistical comparisons were made using the Wilcoxon test. (C) Boxplot comparing the number of rarefied Veillonella counts between the enrollment groups. Statistical comparisons were made using the Wilcoxon test. A summary of the number of infants and samples by group for metabolomics is given in Table 1.
Figure 7
Figure 7
Stool immune marker levels show modest correlation with gut microbiota. (A) Heatmap of repeated measures correlation coefficients between immune markers measured from stool and the most abundant bacterial genera from the same samples (n = 110 stool samples). Only the bacterial genera with at least one significant correlation with an immune marker are displayed (10 genera). Significant correlations are marked with an ‘*’ by the coefficient, with FDR-adjusted p-values < 0.05. (B) Table listing the immune markers used for correlation analysis and their commonly known general functions. (C) Comparison of log10-transformed number of Citrobacter counts by enrollment group and their significance by the Wilcoxon test. A summary of the number of infants and samples by group for immune marker analysis is given in Table 1.

References

    1. Israel EJ. Neonatal necrotizing enterocolitis, a disease of the immature intestinal mucosal barrier. Acta. Paediatr. Suppl. 1994;396:27–32. doi: 10.1111/j.1651-2227.1994.tb13238.x.
    1. Sadeghi K, et al. Immaturity of infection control in preterm and term newborns is associated with Impaired Toll-like receptor signaling. J. Infect. Dis. 2007;195:296–302. doi: 10.1086/509892.
    1. Hunter CJ, Upperman JS, Ford HR, Camerini V. Understanding the susceptibility of the premature infant to necrotizing enterocolitis (NEC) Pediatr. Res. 2008;63:117–123. doi: 10.1203/PDR.0b013e31815ed64c.
    1. Ardissone AN, et al. Meconium microbiome analysis identifies bacteria correlated with premature birth. PLoS ONE. 2014;9:e90784. doi: 10.1371/journal.pone.0090784.
    1. Groer MW, et al. The very low birth weight infant microbiome and childhood health. Birth Defects Res. C Embryo Today. 2015;105:252–264. doi: 10.1002/bdrc.21115.
    1. Pammi M, et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: A systematic review and meta-analysis. Microbiome. 2017;5:31. doi: 10.1186/s40168-017-0248-8.
    1. Patel AL, et al. Longitudinal survey of microbiota in hospitalized preterm very-low-birth-weight infants. J. Pediatr. Gastroenterol. Nutr. 2016;62:292–303. doi: 10.1097/MPG.0000000000000913.
    1. Rozé JC, et al. Assessment of neonatal intensive care unit practices and preterm newborn gut microbiota and 2-year neurodevelopmental outcomes. JAMA Netw. Open. 2020;3:e2018119. doi: 10.1001/jamanetworkopen.2020.18119.
    1. Basso O, Wilcox A. Mortality risk among preterm babies: Immaturity vs. underlying pathology. Epidemiology. 2010;21:521–527. doi: 10.1097/EDE.0b013e3181debe5e.
    1. Tripathi N, Cotten CM, Smith PB. Antibiotic use and misuse in the neonatal intensive care unit. Clin. Perinatol. 2012;39:61–68. doi: 10.1016/j.clp.2011.12.003.
    1. Schulman J, et al. Neonatal intensive care unit antibiotic use. Pediatrics. 2015;135:826–833. doi: 10.1542/peds.2014-3409.
    1. Greenberg RG, et al. Prolonged duration of early antibiotic therapy in extremely premature infants. Pediatr. Res. 2019;85:994–1000. doi: 10.1038/s41390-019-0300-4.
    1. Puopolo KM, Benitz WE, Zaoutis TE, Committee on Fetus and Newborn and Committee on Infectious Diseases Management of neonates born at ≤34 6/7 weeks’ gestation with suspected or proven early-onset bacterial sepsis. Pediatrics. 2018;142:1006.
    1. Fajardo C, Alshaikh B, Harabor A. Prolonged use of antibiotics after birth is associated with increased morbidity in preterm infants with negative cultures. J. Matern.-Fetal Neonatal Med. 2019;32:4060–4066. doi: 10.1080/14767058.2018.1481042.
    1. Samuels N, van de Graaf RA, de Jonge RCJ, Reiss IKM, Vermeulen MJ. Risk factors for necrotizing enterocolitis in neonates: a systematic review of prognostic studies. BMC Pediatr. 2017;17:105. doi: 10.1186/s12887-017-0847-3.
    1. Alganabi, M., Lee, C., Bindi, E., Li, B. & Pierro, A. Recent advances in understanding necrotizing enterocolitis. F1000Res.8, 107 (2019).
    1. Clark RH, Bloom BT, Spitzer AR, Gerstmann DR. Reported medication use in the neonatal intensive care unit: data from a large national data set. Pediatrics. 2006;117:1979–1987. doi: 10.1542/peds.2005-1707.
    1. Hsieh EM, et al. Medication use in the neonatal intensive care unit. Am. J. Perinatol. 2014;31:811–821. doi: 10.1055/s-0033-1361933.
    1. Derrien, M., Alvarez, A.-S. & Vos, W. M. de. The gut microbiota in the first decade of life. Trends Microbiol.27, 997–1010 (2019).
    1. Shao Y, et al. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature. 2019;574:117–121. doi: 10.1038/s41586-019-1560-1.
    1. Stinson LF, Boyce MC, Payne MS, Keelan JA. The not-so-sterile womb: evidence that the human fetus is exposed to bacteria prior to birth. Front. Microbiol. 2019;10:1124. doi: 10.3389/fmicb.2019.01124.
    1. Dardas M, et al. The impact of postnatal antibiotics on the preterm intestinal microbiome. Pediatr. Res. 2014;76:150–158. doi: 10.1038/pr.2014.69.
    1. Gasparrini AJ, et al. Antibiotic perturbation of the preterm infant gut microbiome and resistome. Gut Microbes. 2016;7:443–449. doi: 10.1080/19490976.2016.1218584.
    1. Sun X, Zhuan C, Xiao J, Yao E, Chen L. Impact of postnatal exposure to antibiotics on intestinal microbiome in preterm infants. Chin. J. Perinatal Med. 2018;21:458–464.
    1. Zou Z-H, et al. Prenatal and postnatal antibiotic exposure influences the gut microbiota of preterm infants in neonatal intensive care units. Ann. Clin. Microbiol. Antimicrob. 2018;17:9. doi: 10.1186/s12941-018-0264-y.
    1. Cantey JB, Pyle AK, Wozniak PS, Hynan LS, Sánchez PJ. Early antibiotic exposure and adverse outcomes in preterm, very low birth weight infants. J. Pediatr. 2018;203:62–67. doi: 10.1016/j.jpeds.2018.07.036.
    1. Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med. 2016;8:39. doi: 10.1186/s13073-016-0294-z.
    1. Sherman MP, Zaghouani H, Niklas V. Gut microbiota, the immune system, and diet influence the neonatal gut–brain axis. Pediatr. Res. 2015;77:127–135. doi: 10.1038/pr.2014.161.
    1. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012;10:735–742. doi: 10.1038/nrmicro2876.
    1. Ruoss JL, et al. Routine early antibiotic use in symptomatic preterm neonates (REASON): A pilot randomized controlled trial. J. Pediatr. 2020 doi: 10.1016/j.peds.2020.09.056.
    1. Lindstrom MJ, Bates DM. Newton–Raphson and EM algorithms for linear mixed-effects models for repeated-measures data. J. Am. Stat. Assoc. 1988;83:1014–1022.
    1. Bokulich, N. A. et al. q2-longitudinal: Longitudinal and paired-sample analyses of microbiome data. mSystems3, e00219–18 (2018).
    1. Bolyen E, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 2019;37:852–857. doi: 10.1038/s41587-019-0209-9.
    1. Douglas GM, et al. PICRUSt2: An improved and customizable approach for metagenome inference. bioRxiv. 2020 doi: 10.1101/672295.
    1. Tanaka S, et al. Influence of antibiotic exposure in the early postnatal period on the development of intestinal microbiota. FEMS Immunol. Med. Mic. 2009;56:80–87. doi: 10.1111/j.1574-695X.2009.00553.x.
    1. Greenwood C, et al. Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter. J. Pediatr. 2014;165:23–29. doi: 10.1016/j.jpeds.2014.01.010.
    1. Cong X, et al. Gut microbiome developmental patterns in early life of preterm infants: impacts of feeding and gender. PLoS ONE. 2016;11:e0152751. doi: 10.1371/journal.pone.0152751.
    1. Cong X, et al. Influence of infant feeding type on gut microbiome development in hospitalized preterm infants. Nurs. Res. 2017;66:123–133. doi: 10.1097/NNR.0000000000000208.
    1. Parra-Llorca A, et al. Preterm gut microbiome depending on feeding type: significance of donor human milk. Front. Microbiol. 2018;9:1376. doi: 10.3389/fmicb.2018.01376.
    1. Gregory KE, et al. Influence of maternal breast milk ingestion on acquisition of the intestinal microbiome in preterm infants. Microbiome. 2016;4:68. doi: 10.1186/s40168-016-0214-x.
    1. Kordy K, et al. Contributions to human breast milk microbiome and enteromammary transfer of Bifidobacterium breve. PLoS ONE. 2020;15:e0219633. doi: 10.1371/journal.pone.0219633.
    1. Pannaraj PS, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171:647–654. doi: 10.1001/jamapediatrics.2017.0378.
    1. Forbes JD, et al. Association of exposure to formula in the hospital and subsequent infant feeding practices with gut microbiota and risk of overweight in the first year of life. JAMA Pediatr. 2018;172:e181161. doi: 10.1001/jamapediatrics.2018.1161.
    1. Mueller E, Blaser M. Breast milk, formula, the microbiome and overweight. Nat. Rev. Endocrinol. 2018;14:510–511. doi: 10.1038/s41574-018-0066-5.
    1. Zervos EE, Bass SS, Robson MC, Rosemurgy AS. Fluconazole increases bactericidal activity of neutrophils. J. Trauma. 1996;41:10–14. doi: 10.1097/00005373-199607000-00003.
    1. Rougé C, et al. Fecal calprotectin excretion in preterm infants during the neonatal period. PLoS ONE. 2010;5:e11083. doi: 10.1371/journal.pone.0011083.
    1. Coggins SA, Wynn JL, Weitkamp J-H. Infectious causes of necrotizing enterocolitis. Clin. Perinatol. 2015;42:133–154. doi: 10.1016/j.clp.2014.10.012.
    1. Dobson SRM, Baker CJ. Enterococcal sepsis in neonates: Features by age at onset and occurrence of focal infection. Pediatrics. 1990;85:165–171.
    1. O’Grady NP, et al. Detection of macrophage inflammatory protein (MIP)-1α and MIP-β during experimental endotoxemia and human sepsis. J. Infect. 1999;179:136–141. doi: 10.1086/314559.
    1. Knoop KA, et al. Maternal activation of the EDFR prevents translocation of gut-residing pathogenic Escherichia coli in a model of late-onset neonatal sepsis. PNAS. 2020;117:7941–7949. doi: 10.1073/pnas.1912022117.
    1. Buret A, Olson ME, Gall DG, Hardin JA. Effects of orally administered epidermal growth factor on enteropathogenic Escherichia coli infection in rabbits. Infect. Immunol. 1998;66:4917–4923. doi: 10.1128/IAI.66.10.4917-4923.1998.
    1. Bhutta AT, Cleves MA, Casey PH, Cradock MM, Anand KJS. Cognitive and behavioral outcomes of school-aged children who were born preterm: a meta-analysis. JAMA. 2002;288:728–737. doi: 10.1001/jama.288.6.728.
    1. Counsell SJ, Boardman JP. Differential brain growth in the infant born preterm: Current knowledge and future developments from brain imaging. Semin. Fetal. Neonat. Med. 2005;10:403–410. doi: 10.1016/j.siny.2005.05.003.
    1. Marlow N, Samara M. Neurologic and developmental disability at six years of age after extremely preterm birth. N. Engl. J. Med. 2005;352:9–19. doi: 10.1056/NEJMoa041367.
    1. Sharon G, et al. Specialized metabolites from the microbiome in health and disease. Cell Metab. 2014;20:719–730. doi: 10.1016/j.cmet.2014.10.016.
    1. Kwon SH, et al. GABA, Resting state connectivity and the developing brain. Neonatology. 2014;106:149–155. doi: 10.1159/000362433.
    1. Jin Z, Mendu SK, Birnir B. GABA is an effective immunomodulatory molecule. Amino Acids. 2013;45:87–94. doi: 10.1007/s00726-011-1193-7.
    1. Morland C, et al. Propionate enters GABAergic neurons, inhibits GABA transaminase, causes GABA accumulation and lethargy in a model of propionic acidemia. Biochem J. 2018;475:749–758. doi: 10.1042/BCJ20170814.
    1. Grill JP, Manginot-Dürr C, Schneider F, Ballongue J. Bifidobacteria and probiotic effects: Action of Bifidobacterium species on conjugated bile salts. Curr. Microbiol. 1995;31:23–27. doi: 10.1007/BF00294629.
    1. Jia W, Xie G, Jia W. Bile acid–microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat. Rev. Gastro. Hepat. 2018;15:111–128. doi: 10.1038/nrgastro.2017.119.
    1. Kiriyama Y, Nochi H. The biosynthesis, signaling, and neurological functions of bile acids. Biomolecules. 2019;9:232. doi: 10.3390/biom9060232.
    1. Russell JT, et al. Genetic risk for autoimmunity is associated with distinct changes in the human gut microbiome. Nat. Commun. 2019;10:1–12. doi: 10.1038/s41467-019-11460-x.
    1. Callahan BJ, et al. DADA2: High resolution sample inference from Illumina amplicon data. Nat. Methods. 2016;13:581–583. doi: 10.1038/nmeth.3869.
    1. R Core Team. R: A Language and Environment for Statistical Computing, R Foundation for Statistical Computing, Vienna, Austria. (2020).
    1. Pruesse E, et al. SILVA: A comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res. 2007;35:7188–7196. doi: 10.1093/nar/gkm864.
    1. Glöckner FO, et al. 25 years of serving the community with ribosomal RNA gene reference databases and tools. J. Biotechnol. 2017;261:169–176. doi: 10.1016/j.jbiotec.2017.06.1198.
    1. Yilmaz P, et al. The SILVA and “all-species living tree project (LTP)” taxonomic frameworks. Nucleic Acids Res. 2014;42:D643–D648. doi: 10.1093/nar/gkt1209.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–D596. doi: 10.1093/nar/gks1219.
    1. Stoddard SF, Smith BJ, Hein R, Roller BRK, Schmidt TM. rrnDB: improved tools for interpreting rRNA gene abundance in bacteria and archaea and a new foundation for future development. Nucleic Acids Res. 2015;43:D593–D598. doi: 10.1093/nar/gku1201.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 2007;73:5261–5267. doi: 10.1128/AEM.00062-07.
    1. Chamberlain CA, Hatch M, Garrett TJ. Metabolomic and lipidomic characterization of Oxalobacter formigenes strains HC1 and OxWR by UHPLC-HRMS. Anal. Bioanal. Chem. 2019;411:4807–4818. doi: 10.1007/s00216-019-01639-y.
    1. Chambers MC, et al. A cross-platform toolkit for mass spectrometry and proteomics. Nat. Biotechnol. 2012;30:918–920. doi: 10.1038/nbt.2377.
    1. Pluskal T, Castillo S, Villar-Briones A, Orešič M. MZmine 2: Modular framework for processing, visualizing, and analyzing mass spectrometry-based molecular profile data. BMC Bioinform. 2010;11:395. doi: 10.1186/1471-2105-11-395.
    1. McMurdie PJ, Holmes S. phyloseq: An R package for reproducible interactive analysis and graphics of microbiome census Data. PLoS ONE. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.
    1. Wickham H. ggplot2: Elegant Graphics for Data Analysis. New York: Springer; 2016.
    1. McDonald D, et al. The biological observation matrix (BIOM) format or: How I learned to stop worrying and love the Ome–Ome. Gigascience. 2012;1:1–7. doi: 10.1186/2047-217X-1-7.
    1. Wilcoxon F. Individual comparisons by ranking methods. Biom. Bull. 1945;1:80–83. doi: 10.2307/3001968.
    1. Kruskal WH, Wallis WA. Use of ranks in one-criterion variance analysis. J. Am. Stat. Assoc. 1952;47:583–621. doi: 10.1080/01621459.1952.10483441.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. 1995;57:289–300.
    1. Bakdash JZ, Marusich LR. Repeated measures correlation. Front. Psychol. 2017;8:456. doi: 10.3389/fpsyg.2017.00456.

Source: PubMed

3
Abonner