Microbiome assembly across multiple body sites in low-birthweight infants

Elizabeth K Costello, Erica M Carlisle, Elisabeth M Bik, Michael J Morowitz, David A Relman, Elizabeth K Costello, Erica M Carlisle, Elisabeth M Bik, Michael J Morowitz, David A Relman

Abstract

The purpose of this study was to evaluate the composition and richness of bacterial communities associated with low-birthweight (LBW) infants in relation to host body site, individual, and age. Bacterial 16S rRNA genes from saliva samples, skin swabs, and stool samples collected on postnatal days 8, 10, 12, 15, 18, and 21 from six LBW (five premature) infants were amplified, pyrosequenced, and analyzed within a comparative framework that included analogous data from normal-birthweight (NBW) infants and healthy adults. We found that body site was the primary determinant of bacterial community composition in the LBW infants. However, site specificity depended on postnatal age: saliva and stool compositions diverged over time but were not significantly different until the babies were 15 days old. This divergence was primarily driven by progressive temporal turnover in the distal gut, which proceeded at a rate similar to that of age-matched NBW infants. Neonatal skin was the most adult-like in microbiota composition, while saliva and stool remained the least so. Compositional variation among infants was marked and depended on body site and age. Only the smallest, most premature infant received antibiotics during the study period; this heralded a coexpansion of Pseudomonas aeruginosa and a novel Mycoplasma sp. in the oral cavity of this vaginally delivered, intubated patient. We conclude that concurrent molecular surveillance of multiple body sites in LBW neonates reveals a delayed compositional differentiation of the oral cavity and distal gut microbiota and, in the case of one infant, an abundant, uncultivated oral Mycoplasma sp., recently detected in human vaginal samples.

Importance: Complications of premature birth are the most common cause of neonatal mortality. Colonization by the indigenous microbiota, which begins at delivery, may predispose some high-risk newborns to invasive infection or necrotizing enterocolitis (NEC), and protect others, yet neonatal microbiome dynamics are poorly understood. Here, we present the first cultivation-independent time series tracking microbiota assembly across multiple body sites in a synchronous cohort of hospitalized low-birthweight (LBW) neonates. We take advantage of archived samples and publically available sequence data and compare our LBW infant findings to those from normal-birthweight (NBW) infants and healthy adults. Our results suggest potential windows of opportunity for the dispersal of microbes within and between hosts and support recent findings of substantial baseline spatiotemporal variation in microbiota composition among high-risk newborns.

Figures

FIG 1
FIG  1
Stacked bar plots depicting the relative abundances of the 30 most abundant genus-level taxa in the LBW infants. Taxa were ranked according to their mean abundance across all samples (percentages at right). Ten taxa had mean abundances of >1.00% (percentages in bold type within parentheses). Cs, Cesarean section delivery; V, vaginal delivery.
FIG 2
FIG  2
Unweighted UniFrac-based principal coordinate analysis (PCoA) of LBW infant-associated bacterial communities. Each symbol represents the value for a sample, with the shape of the symbol indicating the infant (infants 1 to 6) and the color indicating the body site. The percentages of variation explained by the plotted principal coordinates (PCo1 and PCo2) are indicated on the axes.
FIG 3
FIG  3
Average unweighted UniFrac distances between LBW infants (present study) and healthy adults (references , and ; see Materials and Methods) for oral, skin surface, and stool microbiota (250 sequences per sample). Values that are significantly different by Tukey’s posthoc tests are indicated by bars and 4 asterisks (P < 0.0001). Error bars represent 95% confidence intervals.
FIG 4
FIG 4
Relationship between neonatal stool microbiota composition and time. (a) Average (95% CI) within-subject, unweighted UniFrac distance plotted against the age difference (lag) in days for LBW (R2 = 0.5) and NBW (R2 = 0.5) infants. Lines indicate best fit linear regressions. The NBW infants were not sampled on day 18; thus, for this analysis, the corresponding age was excluded from the LBW data set. (b) For LBW infants at various ages, average (95% CI) unweighted UniFrac distance compared to a healthy reference cohort (the 21-day-old NBW infants).
FIG 5
FIG  5
Relative abundances of three OTUs belonging to the Mycoplasmataceae from oral samples from extremely LBW baby 3. OTU 15, a novel, uncultivated Mycoplasma sp., is plotted against the upper y axis. OTUs 53 and 144, which are closely related to OTUs from cultivated strains, are plotted against the lower y axis. Expansion of OTU 15 coincided with antibiotic treatment from DOL 13 to 19 (see Table 2 for details). Feedings were delivered via naso- or orogastric tube. For antibiotics, the date range is indicated. The baby’s diet (breast milk or formula) is given for each sample date.

References

    1. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. 2009. Bacterial community variation in human body habitats across space and time. Science 326:1694–1697
    1. Microbiome Human, Project Consortium 2012. Structure, function and diversity of the healthy human microbiome. Nature 486:207–214
    1. Caporaso JG, Lauber CL, Costello EK, Berg-Lyons D, Gonzalez A, Stombaugh J, Knights D, Gajer P, Ravel J, Fierer N, Gordon JI, Knight R. 2011. Moving pictures of the human microbiome. Genome Biol. 12:R50.10.1186/gb-2011-12-5-r50
    1. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R. 2010. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. U. S. A. 107:11971–11975
    1. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. 2007. Development of the human infant intestinal microbiota. PLoS Biol. 5:e177.10.1371/journal.pbio.0050177
    1. Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent LT, Ley RE. 2011. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. U. S. A. 108(Suppl 1):4578–4585
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI. 2012. Human gut microbiome viewed across age and geography. Nature 486:222–227
    1. Capone KA, Dowd SE, Stamatas GN, Nikolovski J. 2011. Diversity of the human skin microbiome early in life. J. Invest. Dermatol. 131:2026–2032
    1. Crielaard W, Zaura E, Schuller AA, Huse SM, Montijn RC, Keijser BJ. 2011. Exploring the oral microbiota of children at various developmental stages of their dentition in the relation to their oral health. BMC Med. Genomics 4:22.10.1186/1755-8794-4-22
    1. Morowitz MJ, Poroyko V, Caplan M, Alverdy J, Liu DC. 2010. Redefining the role of intestinal microbes in the pathogenesis of necrotizing enterocolitis. Pediatrics 125:777–785
    1. Neu J, Walker WA. 2011. Necrotizing enterocolitis. N. Engl. J. Med. 364:255–264
    1. Stoll BJ, Hansen N, Fanaroff AA, Wright LL, Carlo WA, Ehrenkranz RA, Lemons JA, Donovan EF, Stark AR, Tyson JE, Oh W, Bauer CR, Korones SB, Shankaran S, Laptook AR, Stevenson DK, Papile LA, Poole WK. 2002. Late-onset sepsis in very low birth weight neonates: the experience of the NICHD Neonatal Research Network. Pediatrics 110:285–291
    1. Yee WH, Soraisham AS, Shah VS, Aziz K, Yoon W, Lee SK, Canadian Neonatal Network 2012. Incidence and timing of presentation of necrotizing enterocolitis in preterm infants. Pediatrics 129:e298–e304
    1. González-Rivera R, Culverhouse RC, Hamvas A, Tarr PI, Warner BB. 2011. The age of necrotizing enterocolitis onset: an application of Sartwell’s incubation period model. J. Perinatol. 31:519–523
    1. Levy O. 2007. Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat. Rev. Immunol. 7:379–390
    1. Hooper LV, Gordon JI. 2001. Commensal host-bacterial relationships in the gut. Science 292:1115–1118
    1. Sekirov I, Finlay BB. 2009. The role of the intestinal microbiota in enteric infection. J. Physiol. (Lond.) 587:4159–4167
    1. Cho I, Yamanishi S, Cox L, Methé BA, Zavadil J, Li K, Gao Z, Mahana D, Raju K, Teitler I, Li H, Alekseyenko AV, Blaser MJ. 2012. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature 488:621–626
    1. Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, Blumberg RS. 2012. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336:489–493
    1. Carlisle EM, Morowitz MJ. 2011. Pediatric surgery and the human microbiome. J. Pediatr. Surg. 46:577–584
    1. Manzoni P, De Luca D, Stronati M, Jacqz-Aigrain E, Ruffinazzi G, Luparia M, Tavella E, Boano E, Castagnola E, Mostert M, Farina D. 2013. Prevention of nosocomial infections in neonatal intensive care units. Am. J. Perinatol. 30:81–88
    1. Millar MR, Linton CJ, Cade A, Glancy D, Hall M, Jalal H. 1996. Application of 16S rRNA gene PCR to study bowel flora of preterm infants with and without necrotizing enterocolitis. J. Clin. Microbiol. 34:2506–2510
    1. Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, Antonopoulos DA, Chang EB, Claud EC. 2009. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. ISME J. 3:944–954
    1. Mshvildadze M, Neu J, Shuster J, Theriaque D, Li N, Mai V. 2010. Intestinal microbial ecology in premature infants assessed with non-culture-based techniques. J. Pediatr. 156:20–25
    1. Chang JY, Shin SM, Chun J, Lee JH, Seo JK. 2011. Pyrosequencing-based molecular monitoring of the intestinal bacterial colonization in preterm infants. J. Pediatr. Gastroenterol. Nutr. 53:512–519
    1. Mai V, Young CM, Ukhanova M, Wang X, Sun Y, Casella G, Theriaque D, Li N, Sharma R, Hudak M, Neu J. 2011. Fecal microbiota in premature infants prior to necrotizing enterocolitis. PLoS One 6:e20647.10.1371/journal.pone.0020647
    1. Morowitz MJ, Denef VJ, Costello EK, Thomas BC, Poroyko V, Relman DA, Banfield JF. 2011. Strain-resolved community genomic analysis of gut microbial colonization in a premature infant. Proc. Natl. Acad. Sci. U. S. A. 108:1128–1133
    1. Madan JC, Salari RC, Saxena D, Davidson L, O’Toole GA, Moore JH, Sogin ML, Foster JA, Edwards WH, Palumbo P, Hibberd PL. 2012. Gut microbial colonisation in premature neonates predicts neonatal sepsis. Arch. Dis. Child. (Fetal Neonatal Ed.) 97:F456–F462
    1. Sharon I, Morowitz MJ, Thomas BC, Costello EK, Relman DA, Banfield JF. 2013. Time series community genomics analysis reveals rapid shifts in bacterial species, strains, and phage during infant gut colonization. Genome Res. 23:111–120
    1. Normann E, Fahlén A, Engstrand L, Lilja HE. 2013. Intestinal microbial profiles in extremely preterm infants with and without necrotizing enterocolitis. Acta Paediatr. 102:129–136
    1. Mai V, Torrazza RM, Ukhanova M, Wang X, Sun Y, Li N, Shuster J, Sharma R, Hudak ML, Neu J. 2013. Distortions in development of intestinal microbiota associated with late onset sepsis in preterm infants. PLoS One 8:e52876.10.1371/journal.pone.0052876
    1. Morrow AL, Lagomarcino AJ, Schibler KR, Taft DH, Yu Z, Wang B, Altaye M, Wagner M, Gevers D, Ward DV, Kennedy MA, Huttenhower C, Newburg DS. 2013. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. Microbiome 1:13.10.1186/2049-2618-1-13
    1. Keyworth N, Millar MR, Holland KT. 1992. Development of cutaneous microflora in premature neonates. Arch. Dis. Child. 67:797–801
    1. Lozupone C, Knight R. 2005. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 71:8228–8235
    1. Bennet R, Nord CE. 1987. Development of the faecal anaerobic microflora after caesarean section and treatment with antibiotics in newborn infants. Infection 15:332–336
    1. Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, van den Brandt PA, Stobberingh EE. 2006. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics 118:511–521
    1. Martin DH, Zozaya M, Lillis RA, Myers L, Nsuami MJ, Ferris MJ. 2013. Unique vaginal microbiota which include an unknown Mycoplasma-like organism are associated with Trichomonas vaginalis infection. J. Infect. Dis. 207:1922–1931
    1. Mackie RI, Sghir A, Gaskins HR. 1999. Developmental microbial ecology of the neonatal gastrointestinal tract. Am. J. Clin. Nutr. 69:1035S–1045S
    1. Waites KB, Katz B, Schelonka RL. 2005. Mycoplasmas and ureaplasmas as neonatal pathogens. Clin. Microbiol. Rev. 18:757–789
    1. Kuczynski J, Costello EK, Nemergut DR, Zaneveld J, Lauber CL, Knights D, Koren O, Fierer N, Kelley ST, Ley RE, Gordon JI, Knight R. 2010. Direct sequencing of the human microbiome readily reveals community differences. Genome Biol. 11:210.10.1186/gb-2010-11-5-210
    1. Schaedler RW, Dubos R, Costello R. 1965. The development of the bacterial flora in the gastrointestinal tract of mice. J. Exp. Med. 122:59–66
    1. Savage DC. 1977. Microbial ecology of the gastrointestinal tract. Annu. Rev. Microbiol. 31:107–133
    1. Murgas Torrazza R, Neu J. 2011. The developing intestinal microbiome and its relationship to health and disease in the neonate. J. Perinatol. 31(Suppl 1):S29–S34
    1. DiGiulio DB, Romero R, Amogan HP, Kusanovic JP, Bik EM, Gotsch F, Kim CJ, Erez O, Edwin S, Relman DA. 2008. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One 3:e3056.10.1371/journal.pone.0003056
    1. Taylor-Robinson D, Bébéar C. 1997. Antibiotic susceptibilities of mycoplasmas and treatment of mycoplasmal infections. J. Antimicrob. Chemother. 40:622–630
    1. Waites KB, Crouse DT, Cassell GH. 1992. Antibiotic susceptibilities and therapeutic options for Ureaplasma urealyticum infections in neonates. Pediatr. Infect. Dis. J. 11:23–29
    1. de la Cochetiere MF, Piloquet H, des Robert C, Darmaun D, Galmiche JP, Rozé JC. 2004. Early intestinal bacterial colonization and necrotizing enterocolitis in premature infants: the putative role of Clostridium. Pediatr. Res. 56:366–370
    1. Stewart CJ, Marrs EC, Magorrian S, Nelson A, Lanyon C, Perry JD, Embleton ND, Cummings SP, Berrington JE. 2012. The preterm gut microbiota: changes associated with necrotizing enterocolitis and infection. Acta Paediatr. 101:1121–1127
    1. Zoetendal EG, Heilig HG, Klaassens ES, Booijink CC, Kleerebezem M, Smidt H, De Vos WM. 2006. Isolation of DNA from bacterial samples of the human gastrointestinal tract. Nat. Protoc. 1:870–873
    1. Hamady M, Walker JJ, Harris JK, Gold NJ, Knight R. 2008. Error-correcting barcoded primers for pyrosequencing hundreds of samples in multiplex. Nat. Methods 5:235–237
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Peña AG, Goodrich JK, Gordon JI, Huttley GA, Kelley ST, Knights D, Koenig JE, Ley RE, Lozupone CA, McDonald D, Muegge BD, Pirrung M, Reeder J, Sevinsky JR, Turnbaugh PJ, Walters WA, Widmann J, Yatsunenko T, Zaneveld J, Knight R. 2010. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7:335–336
    1. Edgar RC. 2010. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26:2460–2461
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. 2011. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27:2194–2200
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huber T, Dalevi D, Hu P, Andersen GL. 2006. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol. 72:5069–5072
    1. Caporaso JG, Bittinger K, Bushman FD, DeSantis TZ, Andersen GL, Knight R. 2010. PyNAST: a flexible tool for aligning sequences to a template alignment. Bioinformatics 26:266–267
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. 2007. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73:5261–5267
    1. Lane DJ. 1991. 16S/23S rRNA sequencing, p 115–148 In Stackebrandt E, Goodfellow M. (ed), Nucleic acid techniques in bacterial systematics. John Wiley & Sons, Chichester, United Kingdom
    1. Price MN, Dehal PS, Arkin AP. 2010. FastTree 2—approximately maximum-likelihood trees for large alignments. PLoS One 5:e9490.10.1371/journal.pone.0009490
    1. Clarke KR, Gorley RN. 2006. PRIMER v6: user manual/tutorial. PRIMER-E, Plymouth, United Kingdom
    1. DeSantis TZ, Hugenholtz P, Keller K, Brodie EL, Larsen N, Piceno YM, Phan R, Andersen GL. 2006. NAST: a multiple sequence alignment server for comparative analysis of 16S rRNA genes. Nucleic Acids Res. 34:W394–W399
    1. Ludwig W, Strunk O, Westram R, Richter L, Meier H, Kumar Y, Buchner A, Lai T, Steppi S, Jobb G, Förster W, Brettske I, Gerber S, Ginhart AW, Gross O, Grumann S, Hermann S, Jost R, König A, Liss T, Lüssmann R, May M, Nonhoff B, Reichel B, Strehlow R, Stamatakis A, Stuckmann N, Vilbig A, Lenke M, Ludwig T, Bode A, Schleifer KH. 2004. ARB: a software environment for sequence data. Nucleic Acids Res. 32:1363–1371
    1. Stamatakis A, Hoover P, Rougemont J. 2008. A rapid bootstrap algorithm for the RAxML Web servers. Syst. Biol. 57:758–771
    1. Bik EM, Long CD, Armitage GC, Loomer P, Emerson J, Mongodin EF, Nelson KE, Gill SR, Fraser-Liggett CM, Relman DA. 2010. Bacterial diversity in the oral cavity of 10 healthy individuals. ISME J. 4:962–974
    1. Koren O, Goodrich JK, Cullender TC, Spor A, Laitinen K, Bäckhed HK, Gonzalez A, Werner JJ, Angenent LT, Knight R, Bäckhed F, Isolauri E, Salminen S, Ley RE. 2012. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 150:470–480

Source: PubMed

Подписаться