Intestinal dysbiosis in children with short bowel syndrome is associated with impaired outcome

Helene Engstrand Lilja, Hugo Wefer, Niklas Nyström, Yigael Finkel, Lars Engstrand, Helene Engstrand Lilja, Hugo Wefer, Niklas Nyström, Yigael Finkel, Lars Engstrand

Abstract

Background: The composition of the intestinal microbiota seems to be an important factor in determining the clinical outcome in children with short bowel syndrome (SBS). Alterations in the microbiota may result in serious complications such as small bowel bacterial overgrowth (SBBO) and intestinal mucosal inflammation that lead to prolonged parenteral nutrition (PN) dependency with subsequently increased risk of liver failure and sepsis. To date, there are no reported mappings of the intestinal microbiome in children with SBS. Here, we present the first report on the intestinal microbial community profile in children with SBS.

Findings: The study includes children diagnosed with SBS in the neonatal period. Healthy siblings served as controls. Fecal samples were collected, and microbial profiles were analyzed by using 16S rRNA gene sequencing on the Illumina MiSeq platform. We observed a pronounced microbial dysbiosis in children with SBS on PN treatment with an increased and totally dominating relative abundance of Enterobacteriacae in four out of five children compared to children with SBS weaned from PN and healthy siblings.

Conclusions: The overall decreased bacterial diversity in children with SBS is consistent with intestinal microbiome mappings in inflammatory bowel diseases such as Crohn's disease and necrotizing enterocolitis in preterm infants. Our findings indicate that intestinal dysbiosis in children with SBS is associated with prolonged PN dependency.

Keywords: Bacterial diversity; Dysbiosis; Gut microbiota; Short bowel syndrome.

Figures

Figure 1
Figure 1
Shannon diversity index in children with SBS still on PN compared to children weaned from PN.
Figure 2
Figure 2
Microbial communities in children with SBS on PN (1A, 3A, 8A, 9A, 12A), SBS children weaned from PN (2A, 4A, 11A, 13A, 16A, 18A), and siblings (2C1, 2C2, 11C1, 11C2, 12C, 13C1, 13C2). The figure is showing the relative abundance of the 19 most common taxonomic families that accounts for at least 84% of the abundance in all samples.
Figure 3
Figure 3
Shannon diversity index in children with SBS on PN (1A, 3A, 8A, 9A, 12A), SBS children weaned from PN (2A, 4A, 11A, 13A, 16A, 18A), and siblings (2C1, 2C2, 11C1, 11C2, 12C, 13C1, 13C2).
Figure 4
Figure 4
PCoA plot describing unweighted UniFrac distance between samples. Pairwise distances between all samples are projected onto a two-dimensional space where the axis PC1 describes the highest degree of variation. Samples that are clustered closely together are thus considered to share a larger proportion of the phylogenetic tree in comparison to samples that are more separated.

References

    1. Gutierrez IM, Kang KH, Jaksic T. Neonatal short bowel syndrome. Semin Fetal Neonatal Med. 2011;16:157–63. doi: 10.1016/j.siny.2011.02.001.
    1. Goulet O, Joly F. Intestinal microbiota in short bowel syndrome. Gastroenterol Clin Biol. 2010;34:S37–43. doi: 10.1016/S0399-8320(10)70019-1.
    1. Kaufman SS, Loseke CA, Lupo JV, Young RJ, Murray ND, Pinch LW, et al. Influence of bacterial overgrowth and intestinal inflammation on duration of parenteral nutrition in children with short bowel syndrome. J Pediatr. 1997;131:356–61. doi: 10.1016/S0022-3476(97)80058-3.
    1. Vanderhoof JA, Langnas AN. Short-bowel syndrome in children and adults. Gastroenterology. 1997;113:1767–78. doi: 10.1053/gast.1997.v113.pm9352883.
    1. Sigalet D, Boctor D, Brindle M, Lam V, Robertson M. Elements of successful intestinal rehabilitation. J Pediatr Surg. 2011;46:150–6. doi: 10.1016/j.jpedsurg.2010.09.083.
    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;486:222–7.
    1. Winter SE, Bäumler AJ. Dysbiosis in the inflamed intestine: chance favors the prepared microbe. Gut Microbes. 2014;5:71–3. doi: 10.4161/gmic.27129.
    1. Normann E, Fahlén A, Engstrand L, Lilja HE. Intestinal microbial profiles in extremely preterm infants with and without necrotizing enterocolitis. Acta Paediatr. 2013;102:129–36. doi: 10.1111/apa.12059.
    1. Kim HB, Fauza D, Garza J, Oh J-T, Nurko S, Jaksic T. Serial transverse enteroplasty (STEP): a novel bowel lengthening procedure. J Pediatr Surg. 2003;38:425–9. doi: 10.1053/jpsu.2003.50073.
    1. Hugerth LW, Wefer HA, Lundin S, Jakobsson HE, Lindberg M, Rodin S, et al. DegePrime, a program for degenerate primer design for broad-taxonomic-range pcr in microbial ecology studies. Appl Environ Microbiol. 2014;80:5116–23. doi: 10.1128/AEM.01403-14.
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7:335–6. doi: 10.1038/nmeth.f.303.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatic. 2010;26:2460–1. doi: 10.1093/bioinformatics/btq461.
    1. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, et al. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut. 2014;63:559–66. doi: 10.1136/gutjnl-2012-303249.
    1. Gutierrez IM, Kang KH, Calvert CE, Johnson VM, Zurakowski D, Kamin D, et al. Risk factors for small bowel bacterial overgrowth and diagnostic yield of duodenal aspirates in children with intestinal failure: a retrospective review. J Pediatr Surg. 2012;47:1150–4. doi: 10.1016/j.jpedsurg.2012.03.019.
    1. Spees AM, Lopez CA, Kingsbury DD, Winter SE, Bäumler AJ. Colonization resistance: battle of the bugs or Ménage à Trois with the host? PLoS Pathog. 2013;9(11) doi: 10.1371/journal.ppat.1003730.
    1. Lacaille F, Gupte G, Colomb V, D’Antiga L, Hartman C, Hojsak I, et al. Intestinal failure-associated liver disease: a position paper of the ESPGHAN working group of intestinal failure and intestinal transplantation. J Pediatr Gastroenterol Nutr. 2015;60:272–83. doi: 10.1097/MPG.0000000000000586.
    1. Walker AW, Sanderson JD, Churcher C, Parkes GC, Hudspith BN, Rayment N, et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2011;11:7. doi: 10.1186/1471-2180-11-7.
    1. Lapthorne S, Pereira-Fantini PM, Fouhy F, Wilson G, Thomas SL, Dellios NL, et al. Gut microbial diversity is reduced and is associated with colonic inflammation in a piglet model of short bowel syndrome. Gut Microbes. 2013;4:212–21. doi: 10.4161/gmic.24372.
    1. Smits LP, Bouter KE, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology. 2013;145:946–53. doi: 10.1053/j.gastro.2013.08.058.
    1. Damman CJ, Miller SI, Surawicz CM, Zisman TL. The microbiome and inflammatory bowel disease: is there a therapeutic role for fecal microbiota transplantation? Am J Gastroenterol. 2012;107:1452–9. doi: 10.1038/ajg.2012.93.
    1. Baron TH, Kozarek RA. Fecal microbiota transplant: we know its history, but can we predict its future? Mayo Clin Proc. 2013;88:782–5. doi: 10.1016/j.mayocp.2013.06.007.

Source: PubMed

3
Suscribir