Antibiotic-driven intestinal dysbiosis in pediatric short bowel syndrome is associated with persistently altered microbiome functions and gut-derived bloodstream infections

Robert Thänert, Anna Thänert, Jocelyn Ou, Adam Bajinting, Carey-Ann D Burnham, Holly J Engelstad, Maria E Tecos, I Malick Ndao, Carla Hall-Moore, Colleen Rouggly-Nickless, Mike A Carl, Deborah C Rubin, Nicholas O Davidson, Phillip I Tarr, Barbara B Warner, Gautam Dantas, Brad W Warner, Robert Thänert, Anna Thänert, Jocelyn Ou, Adam Bajinting, Carey-Ann D Burnham, Holly J Engelstad, Maria E Tecos, I Malick Ndao, Carla Hall-Moore, Colleen Rouggly-Nickless, Mike A Carl, Deborah C Rubin, Nicholas O Davidson, Phillip I Tarr, Barbara B Warner, Gautam Dantas, Brad W Warner

Abstract

Surgical removal of the intestine, lifesaving in catastrophic gastrointestinal disorders of infancy, can result in a form of intestinal failure known as short bowel syndrome (SBS). Bloodstream infections (BSIs) are a major challenge in pediatric SBS management. BSIs require frequent antibiotic therapy, with ill-defined consequences for the gut microbiome and childhood health. Here, we combine serial stool collection, shotgun metagenomic sequencing, multivariate statistics and genome-resolved strain-tracking in a cohort of 19 patients with surgically-induced SBS to show that antibiotic-driven intestinal dysbiosis in SBS enriches for persistent intestinal colonization with BSI causative pathogens in SBS. Comparing the gut microbiome composition of SBS patients over the first 4 years of life to 19 age-matched term and 18 preterm controls, we find that SBS gut microbiota diversity and composition was persistently altered compared to controls. Commensals including Ruminococcus, Bifidobacterium, Eubacterium, and Clostridium species were depleted in SBS, while pathobionts (Enterococcus) were enriched. Integrating clinical covariates with gut microbiome composition in pediatric SBS, we identified dietary and antibiotic exposures as the main drivers of these alterations. Moreover, antibiotic resistance genes, specifically broad-spectrum efflux pumps, were at a higher abundance in SBS, while putatively beneficial microbiota functions, including amino acid and vitamin biosynthesis, were depleted. Moreover, using strain-tracking we found that the SBS gut microbiome harbors BSI causing pathogens, which can persist intestinally throughout the first years of life. The association between antibiotic-driven gut dysbiosis and enrichment of intestinal pathobionts isolated from BSI suggests that antibiotic treatment may predispose SBS patients to infection. Persistence of pathobionts and depletion of beneficial microbiota and functionalities in SBS highlights the need for microbiota-targeted interventions to prevent infection and facilitate intestinal adaptation.

Keywords: Short bowel syndrome; antibiotics; bloodstream infections; functional profiling; intestinal dysbiosis; microbiota; shotgun metagenomics; strain-tracking.

Conflict of interest statement

No potential conflict of interest was reported by the author(s).

Figures

Figure 1.
Figure 1.
Taxonomic composition of the SBS microbiota compared to preterm and term controls. (a) Shannon diversity indices of the gut microbiota of SBS patients (salmon), preterm (purple) and term controls (teal) by year of life (n = 159). Loess regression lines with 95% confidence interval shading are drawn. All P-values are two-tailed, from longitudinal maximum-likelihood GLMMs Tukey-adjusted for multiple comparisons. (b) Boxplot of Shannon diversity of SBS gut microbiota with or without exposure to antibiotics in the month prior to sampling (n = 51, P = .042, longitudinal maximum-likelihood GLMM FDR corrected). (c) Principal Coordinate Analysis (PCoA) plot of species based on the Bray–Curtis dissimilarity index for all samples (n = 159), colored by day of life. (d) Species enriched (salmon) or depleted (teal) in SBS patients compared to term controls as determined via MaAsLin2. (Top) Number of species significantly depleted or enriched in SBS or term controls plotted against determined binned effect sizes. (Bottom) Top 20% of species depleted or enriched in SBS patients compared to term controls selected based on determined effect size
Figure 1.
Figure 1.
Taxonomic composition of the SBS microbiota compared to preterm and term controls. (a) Shannon diversity indices of the gut microbiota of SBS patients (salmon), preterm (purple) and term controls (teal) by year of life (n = 159). Loess regression lines with 95% confidence interval shading are drawn. All P-values are two-tailed, from longitudinal maximum-likelihood GLMMs Tukey-adjusted for multiple comparisons. (b) Boxplot of Shannon diversity of SBS gut microbiota with or without exposure to antibiotics in the month prior to sampling (n = 51, P = .042, longitudinal maximum-likelihood GLMM FDR corrected). (c) Principal Coordinate Analysis (PCoA) plot of species based on the Bray–Curtis dissimilarity index for all samples (n = 159), colored by day of life. (d) Species enriched (salmon) or depleted (teal) in SBS patients compared to term controls as determined via MaAsLin2. (Top) Number of species significantly depleted or enriched in SBS or term controls plotted against determined binned effect sizes. (Bottom) Top 20% of species depleted or enriched in SBS patients compared to term controls selected based on determined effect size
Figure 2.
Figure 2.
Functional composition of the SBS microbiota throughout the first years of life. (a) Principal Coordinate Analysis (PCoA) plot of functional pathway abundance based on the Bray–Curtis dissimilarity index for all samples (n = 159), colored by day of life. (b) Pathways significantly enriched (salmon) or depleted (teal) in SBS patients aggregated into functional categories based on MetaCyc hierarchy compared to term controls as determined via MaAsLin2. (Left and right) Number of pathways significantly depleted or enriched in SBS or term controls within each functional category plotted against determined binned effect sizes. (Middle) Sum of all depleted or enriched pathways within each functional category grouped by depletion and enrichment status in SBS patients compared to term controls. (c) Amino acid synthesis or degradation pathways significantly enriched (salmon) or depleted (teal) in SBS patients compared to term controls as determined via MaAsLin2
Figure 2.
Figure 2.
Functional composition of the SBS microbiota throughout the first years of life. (a) Principal Coordinate Analysis (PCoA) plot of functional pathway abundance based on the Bray–Curtis dissimilarity index for all samples (n = 159), colored by day of life. (b) Pathways significantly enriched (salmon) or depleted (teal) in SBS patients aggregated into functional categories based on MetaCyc hierarchy compared to term controls as determined via MaAsLin2. (Left and right) Number of pathways significantly depleted or enriched in SBS or term controls within each functional category plotted against determined binned effect sizes. (Middle) Sum of all depleted or enriched pathways within each functional category grouped by depletion and enrichment status in SBS patients compared to term controls. (c) Amino acid synthesis or degradation pathways significantly enriched (salmon) or depleted (teal) in SBS patients compared to term controls as determined via MaAsLin2
Figure 3.
Figure 3.
Resistome composition in SBS throughout the first years of life. (a) ARG abundance measured in RPKM within the microbiome of SBS patients (salmon), preterm (purple) and term controls (teal) by year of life (n = 159). Loess regression lines with 95% confidence interval shading are drawn. All P-values are two-tailed, from longitudinal maximum-likelihood GLMMs Tukey-adjusted for multiple comparisons. (b) Boxplot of ARG abundance measured in RPKM within the microbiome of SBS patients with or without exposure to enteral nutrition at sampling (n = 51, P = .011, longitudinal maximum-likelihood GLMM). (c) Principal Coordinate Analysis (PCoA) plot of ARG abundance profiles based on the Bray–Curtis dissimilarity index for all samples (n = 159), colored by day of life
Figure 3.
Figure 3.
Resistome composition in SBS throughout the first years of life. (a) ARG abundance measured in RPKM within the microbiome of SBS patients (salmon), preterm (purple) and term controls (teal) by year of life (n = 159). Loess regression lines with 95% confidence interval shading are drawn. All P-values are two-tailed, from longitudinal maximum-likelihood GLMMs Tukey-adjusted for multiple comparisons. (b) Boxplot of ARG abundance measured in RPKM within the microbiome of SBS patients with or without exposure to enteral nutrition at sampling (n = 51, P = .011, longitudinal maximum-likelihood GLMM). (c) Principal Coordinate Analysis (PCoA) plot of ARG abundance profiles based on the Bray–Curtis dissimilarity index for all samples (n = 159), colored by day of life
Figure 4.
Figure 4.
Gut-persisting pathobionts can cause repeated episodes of BSI in SBS. (a) E. faecalis persists in patient SBS 07 causing two BSI over 3 years of life. (Top) Relative abundance of E. faecalis in stools (red) by year of life. Other species are depicted in gray. Schematic of sample collection and BSI events is shown on top. (Bottom) Phylogenetic relatedness of BSI isolates and metagenomic strains based on core SNPs as assessed by StrainSifter. Branch tip colors indicate BSI isolates (red), SBS (brown) and preterm (peach), or term (teal) stools. (b) K. pneumoniae found in a stool of patient SBS 01 concurrently causes a BSI. (Top) Relative abundance of K. pneumoniae in stools (green) by year of life. Other species are depicted in gray. Schematic of sample collection and BSI events is shown on top. (Bottom) Phylogenetic relatedness of BSI isolates and metagenomic strains based on core SNPs as assessed by StrainSifter. Branch tip colors indicate BSI isolates (red), SBS (brown) and preterm (peach), or term (teal) stools
Figure 4.
Figure 4.
Gut-persisting pathobionts can cause repeated episodes of BSI in SBS. (a) E. faecalis persists in patient SBS 07 causing two BSI over 3 years of life. (Top) Relative abundance of E. faecalis in stools (red) by year of life. Other species are depicted in gray. Schematic of sample collection and BSI events is shown on top. (Bottom) Phylogenetic relatedness of BSI isolates and metagenomic strains based on core SNPs as assessed by StrainSifter. Branch tip colors indicate BSI isolates (red), SBS (brown) and preterm (peach), or term (teal) stools. (b) K. pneumoniae found in a stool of patient SBS 01 concurrently causes a BSI. (Top) Relative abundance of K. pneumoniae in stools (green) by year of life. Other species are depicted in gray. Schematic of sample collection and BSI events is shown on top. (Bottom) Phylogenetic relatedness of BSI isolates and metagenomic strains based on core SNPs as assessed by StrainSifter. Branch tip colors indicate BSI isolates (red), SBS (brown) and preterm (peach), or term (teal) stools

References

    1. Wales PW, De Silva N, Kim J, Lecce L, To T, Moore A.. Neonatal short bowel syndrome: population-based estimates of incidence and mortality rates. J Pediatr Surg. 2004;39(5):690–15. doi:10.1016/j.jpedsurg.2004.01.036.
    1. Chandra R, Kesavan A. Current treatment paradigms in pediatric short bowel syndrome. Clin J Gastroenterol. 2018;11(2):103–112. doi:10.1007/s12328-017-0811-7.
    1. Amin SC, Pappas C, Iyengar H, Maheshwari A. Short bowel syndrome in the NICU [Internet]. Clin Perinatol. 2013;40(1):53–68. doi:10.1016/j.clp.2012.12.003.
    1. Merritt RJ, Cohran V, Raphael BP, Sentongo T, Volpert D, Warner BW, Goday PS. Intestinal rehabilitation programs in the management of pediatric intestinal failure and short bowel syndrome. J Pediatr Gastroenterol Nutr. 2017;65(5):588–596. doi:10.1097/MPG.0000000000001722.
    1. Cole CR, Frem JC, Schmotzer B, Gewirtz AT, Meddings JB, Gold BD, Ziegler TR. The rate of bloodstream infection is high in infants with short bowel syndrome: relationship with small bowel bacterial overgrowth, enteral feeding, and inflammatory and immune responses. J Pediatr. 2010;156(6):941–947.e1. doi:10.1016/j.jpeds.2009.12.008.
    1. Duro D, Kamin D, Duggan C. Overview of pediatric short bowel syndrome. J Pediatr Gastroenterol Nutr. 2008;47(Suppl 1):S33–S36. doi:10.1097/MPG.0b013e3181819007.
    1. Bohm M, Siwiec RM, Wo JM. Diagnosis and management of small intestinal bacterial overgrowth. Nutr Clin Prac. 2013;28(3):289–299. doi:10.1177/0884533613485882.
    1. Cahova M, Bratova M, Wohl P. Parenteral nutrition-associated liver disease: the role of the gut microbiota. Nutrients. 2017;9(9):1–19. doi:10.3390/nu9090987.
    1. Hukkinen M, Mutanen A, Pakarinen MP. Small bowel dilation in children with short bowel syndrome is associated with mucosal damage, bowel-derived bloodstream infections, and hepatic injury. Surg (United States). 2017;162:670–679.
    1. Budinska E, Gojda J, Heczkova M, Bratova M, Dankova H, Wohl P, Bastova H, Lanska V, Kostovcik M, Dastych M, et al. Microbiome and metabolome profiles associated with different types of short bowel syndrome: implications for treatment. J Parenter Enter Nutr. 2020;44(1):105–118. doi:10.1002/jpen.1595.
    1. Wang P, Wang Y, Lu L, Yan W, Tao Y, Zhou K, Jia J, Cai W. Alterations in intestinal microbiota relate to intestinal failure-associated liver disease and central line infections. J Pediatr Surg. 2017;52(8):1318–1326. doi:10.1016/j.jpedsurg.2017.04.020.
    1. Engstrand Lilja H, Wefer H, Nyström N, Finkel Y, Engstrand L. Intestinal dysbiosis in children with short bowel syndrome is associated with impaired outcome. Microbiome. 2015;3(1):18. doi:10.1186/s40168-015-0084-7.
    1. Piper HG, Coughlin LA, Nguyen V, Channabasappa N, Koh AY. A comparison of small bowel and fecal microbiota in children with short bowel syndrome. J Pediatr Surg. 2020;55(5):878–882. doi:10.1016/j.jpedsurg.2020.01.032.
    1. Piper HG, Fan D, Coughlin LA, Ho EX, McDaniel MM, Channabasappa N, Kim J, Kim M, Zhan X, Xie Y, et al. Severe gut microbiota dysbiosis is associated with poor growth in patients with short bowel syndrome. J Parenter Enter Nutr. 2017;41(7):1202–1212. doi:10.1177/0148607116658762.
    1. Channabasappa N, Girouard S, Nguyen V, Piper H. Enteral nutrition in pediatric short-bowel syndrome. Nutr Clin Pract. 2020;35(5):848–854. doi:10.1002/ncp.10565.
    1. Gibson MK, Wang B, Ahmadi S, Burnham C-AD, Tarr PI, Warner BB, Dantas G. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat Microbiol. 2016;1(4):16024. doi:10.1038/nmicrobiol.2016.24.
    1. Gasparrini AJ, Wang B, Sun X, Kennedy EA, Hernandez-Leyva A, Ndao IM, Tarr PI, Warner BB, Dantas G. Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nat Microbiol. 2019;4(12):2285–2297. 10.1038/s41564-019-0550-2
    1. Carl MA, Malick Ndao I, Springman AC, Manning SD, Johnson JR, Johnston BD, Burnham C-AD, Weinstock ES, Weinstock GM, Wylie TN, et al. Sepsis from the gut: the enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. 2014.
    1. Tamburini FB, Andermann TM, Tkachenko E, Senchyna F, Banaei N, Bhatt AS. Precision identification of diverse bloodstream pathogens in the gut microbiome. Nat Med. 2018;24(12):1809–1814. doi:10.1038/s41591-018-0202-8.
    1. DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, Turbett S, Chung RT, Chen Y-B, Hohmann EL. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381(21):2043–2050. doi:10.1056/NEJMoa1910437.
    1. Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The human microbiome and child growth – first 1000 days and beyond. Trends Microbiol. 2019;27(2):131–147. doi:10.1016/j.tim.2018.09.008.
    1. Schall KA, Thornton ME, Isani M, Holoyda KA, Hou X, Lien C-L, Grubbs BH, Grikscheit TC. Short bowel syndrome results in increased gene expression associated with proliferation, inflammation, bile acid synthesis and immune system activation: RNA sequencing a zebrafish SBS model. BMC Genomics. 2017;18(1):23. doi:10.1186/s12864-016-3433-4.
    1. Marchix J, Goddard G, Helmrath MA. Host-gut microbiota crosstalk in intestinal adaptation. CMGH. 2018;6(2):149–162. doi:10.1016/j.jcmgh.2018.01.024.
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi:10.1038/nature11053.
    1. La Rosa PS, Warner BB, Zhou Y, Weinstock GM, Sodergren E, Hall-Moore CM, Stevens HJ, Bennett WE, Shaikh N, Linneman LA, et al. Patterned progression of bacterial populations in the premature infant gut. Proc Natl Acad Sci U S A. 2014;111(34):12522–12527. doi:10.1073/pnas.1409497111.
    1. Mukhopadhyay B, Bourne C, Versalovic J, Engevik MA, Morra CN, Röth D, Engevik K, Spinler JK, Devaraj S, Crawford SE, et al. Microbial metabolic capacity for intestinal folate production and modulation of host folate receptors. Front Microbiol. 2019;10:2305. doi:10.3389/fmicb.2019.02305.
    1. Krautkammer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol. 2021;19:77-94. 10.1038/s41579-020-0438-4
    1. Agus A, Clément K, Sokol H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut. 2020. Jun 1;70(6):1174–1182. doi:10.1136/gutjnl-2020-323071.
    1. Zhou H, Yu B, Gao J, Htoo JK, Chen D. Regulation of intestinal health by branched-chain amino acids. Anim Sci J. 2018;89(1):3–11. doi:10.1111/asj.12937.
    1. Ren M, Zhang SH, Zeng XF, Liu H, Qiao SY. Branched-chain amino acids are beneficial to maintain growth performance and intestinal immune-related function in weaned piglets fed protein restricted diet. Asian-Australasian J Anim Sci. 2015;28(12):1742–1750. doi:10.5713/ajas.14.0131.
    1. Lapthorne S, Pereira-Fantini PM, Fouhy F, Wilson G, Thomas SL, Dellios NL, Scurr M, O’Sullivan O, Paul Ross R, Stanton C, 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(3):212–221. doi:10.4161/gmic.24372.
    1. Cani PD, Osto M, Geurts L, Everard A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes. 2012;3(4):279. doi:10.4161/gmic.19625.
    1. Velasquez OR, Place AR, Tso P, Crissinger KD. Developing intestine is injured during absorption of oleic acid but not its ethyl ester. J Clin Invest. 1994;93(2):479–485. doi:10.1172/JCI116996.
    1. Wisniewski PJ, Dowden RA, Campbell SC. Role of dietary lipids in modulating inflammation through the gut microbiota. Nutrients. 2019;11(1):117. doi:10.3390/nu11010117.
    1. Rowan-Nash AD, Araos R, D’Agata EMC, Belenky P. Antimicrobial resistance gene prevalence in a population of patients with advanced dementia is related to specific pathobionts. iScience. 2020;23(3):100905. doi:10.1016/j.isci.2020.100905.
    1. Engelstad HJ, Barron L, Moen J, Wylie TN, Wylie K, Rubin DC, Davidson N, Cade WT, Warner BB, Warner BW. Remnant small bowel length in pediatric short bowel syndrome and the correlation with intestinal dysbiosis and linear growth. J Am Coll Surg. 2018;227(4):439–449. doi:10.1016/j.jamcollsurg.2018.07.657.
    1. Kelly MS, Ward DV, Severyn CJ, Arshad M, Heston SM, Jenkins K, Martin PL, McGill L, Stokhuyzen A, Bhattarai SK, et al. Gut colonization preceding mucosal barrier injury bloodstream infection in pediatric hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant. 2019;25(11):2274–2280. doi:10.1016/j.bbmt.2019.07.019.
    1. Vanderhoof JA, Young RJ, Murray N, Kaufman SS. Treatment strategies for small bowel bacterial overgrowth in short bowel syndrome. J Pediatr Gastroenterol Nutr. 1998;27(2):155–160. doi:10.1097/00005176-199808000-00005.
    1. Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–662. doi:10.1038/s41586-019-1237-9.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120. doi:10.1093/bioinformatics/btu170.
    1. Schmieder R, Edwards R, Rodriguez-Valera F. Fast identification and removal of sequence contamination from genomic and metagenomic datasets. PLoS One. 2011;6(3):e17288. doi:10.1371/journal.pone.0017288.
    1. Truong DT, Franzosa EA, Tickle TL, Scholz M, Weingart G, Pasolli E, Tett A, Huttenhower C, Segata N. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods. 2015;12(10):902–903. doi:10.1038/nmeth.3589.
    1. Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M, Weingart G, Lipson KS, Knight R, Caporaso JG, Segata N, et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods. 2018;15(11):962–968. doi:10.1038/s41592-018-0176-y.
    1. Kaminski J, Gibson MK, Franzosa EA, Segata N, Dantas G, Huttenhower C, Noble WS. High-specificity targeted functional profiling in microbial communities with ShortBRED. PLOS Comput Biol. 2015;11(12):e1004557. doi:10.1371/journal.pcbi.1004557.
    1. Mallick H, Rahnavard A, McIver LJ, Ma S, Zhang Y, Tickle TL, Weingart G, Ren B, Schwager EH, Thompson KN, et al. Multivariable association discovery in population-scale meta-omics studies 3. bioRxiv. 2021;2021(1):20.427420.
    1. Baumann-Dudenhoeffer AM, D’Souza AW, Tarr PI, Warner BB, Dantas G. Infant diet and maternal gestational weight gain predict early metabolic maturation of gut microbiomes. Nat Med. 2018;24(12):1822–1829. doi:10.1038/s41591-018-0216-2.
    1. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, Lesin VM, Nikolenko SI, Pham S, Prjibelski AD, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19(5):455–477. doi:10.1089/cmb.2012.0021.
    1. Parks DH, Imelfort M, Skennerton CT, Hugenholtz P, Tyson GW. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res. 2015;25(7):1043–1055. doi:10.1101/gr.186072.114.
    1. Gurevich A, Saveliev V, Vyahhi N, Tesler G. QUAST: quality assessment tool for genome assemblies. Bioinformatics. 2013;29(8):1072–1075. doi:10.1093/bioinformatics/btt086.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9(4):357–359. doi:10.1038/nmeth.1923.

Source: PubMed

Подписаться