Multi-Compartment Profiling of Bacterial and Host Metabolites Identifies Intestinal Dysbiosis and Its Functional Consequences in the Critically Ill Child

Anisha Wijeyesekera, Josef Wagner, Marcus De Goffau, Sarah Thurston, Adilson Rodrigues Sabino, Sara Zaher, Deborah White, Jenna Ridout, Mark J Peters, Padmanabhan Ramnarayan, Ricardo G Branco, M Estee Torok, Frederic Valla, Rosan Meyer, Nigel Klein, Gary Frost, Julian Parkhill, Elaine Holmes, Nazima Pathan, Anisha Wijeyesekera, Josef Wagner, Marcus De Goffau, Sarah Thurston, Adilson Rodrigues Sabino, Sara Zaher, Deborah White, Jenna Ridout, Mark J Peters, Padmanabhan Ramnarayan, Ricardo G Branco, M Estee Torok, Frederic Valla, Rosan Meyer, Nigel Klein, Gary Frost, Julian Parkhill, Elaine Holmes, Nazima Pathan

Abstract

Objectives: Adverse physiology and antibiotic exposure devastate the intestinal microbiome in critical illness. Time and cost implications limit the immediate clinical potential of microbial sequencing to identify or treat intestinal dysbiosis. Here, we examined whether metabolic profiling is a feasible method of monitoring intestinal dysbiosis in critically ill children.

Design: Prospective multicenter cohort study.

Setting: Three U.K.-based PICUs.

Patients: Mechanically ventilated critically ill (n = 60) and age-matched healthy children (n = 55).

Interventions: Collection of urine and fecal samples in children admitted to the PICU. A single fecal and urine sample was collected in healthy controls.

Measurements and main results: Untargeted and targeted metabolic profiling using 1H-nuclear magnetic resonance spectroscopy and liquid chromatography-mass spectrometry or urine and fecal samples. This was integrated with analysis of fecal bacterial 16S ribosomal RNA profiles and clinical disease severity indicators. We observed separation of global urinary and fecal metabolic profiles in critically ill compared with healthy children. Urinary excretion of mammalian-microbial co-metabolites hippurate, 4-cresol sulphate, and formate were reduced in critical illness compared with healthy children. Reduced fecal excretion of short-chain fatty acids (including butyrate, propionate, and acetate) were observed in the patient cohort, demonstrating that these metabolites also distinguished between critical illness and health. Dysregulation of intestinal bile metabolism was evidenced by increased primary and reduced secondary fecal bile acid excretion. Fecal butyrate correlated with days free of intensive care at 30 days (r = 0.38; p = 0.03), while urinary formate correlated inversely with vasopressor requirement (r = -0.2; p = 0.037).

Conclusions: Disruption to the functional activity of the intestinal microbiome may result in worsening organ failure in the critically ill child. Profiling of bacterial metabolites in fecal and urine samples may support identification and treatment of intestinal dysbiosis in critical illness.

Figures

Figure 1.
Figure 1.
Urinary and fecal 1H-nuclear magnetic resonance (1H-NMR) global metabolic profiles of critically ill and healthy children. A, Unsupervised principal components analysis (PCA) scores plot of admission urine samples from age-matched critically ill (red) and healthy (blue) children. R2 = 0.16, Q2 = 0.09. B, Supervised Orthogonal Projections to Latent Structures Discriminant Analysis (O-PLS-DA) loadings line plot. Urinary metabolites higher in critically ill children (up) compared with age-matched healthy children (down). The color bar indicates the correlation coefficient (R2) (i.e., the redder the peak, the higher the correlation). R2Y = 0.89, Q2Y = 0.80. C, Unsupervised PCA scores plot of fecal samples from age-matched critically ill (red, n = 27) and healthy control (blue, n = 41) samples. R2 = 0.23, Q2 = 0.08. D, Supervised O-PLS-DA loadings line plot. Fecal metabolites higher in critically ill children (up) compared with age-matched healthy children (down). R2Y = 0.96, Q2Y = 0.85. R2Y = variance explained, Q2Y= predictive ability. a.u = arbitrary units, N-AG = n-acetylglucosamine, NMNA = n-nethylnicotinamide.
Figure 2.
Figure 2.
Fecal liquid chromatography-mass spectrometry bile acid (BA) profiles of critically ill and healthy children. A, Unsupervised principal components analysis scores plot of critically ill (red) and healthy control (blue) samples. R2 = 0.47, Q2 = 0.16. B, Changes in the metabolism of BAs in critically ill (in red) compared with healthy children (in blue) illustrate accumulation of primary BAs and a reduction in lithocholic acid in critically ill children. 5β-CA-3β, 12a-diol = 5β-cholanic acid-3β, 12a-diol, FDR 2.952E-6, 23 nor 5β- CA-3α, 12a-diol = 23-nor-5b-cholanic acid-3a, 12a-diol, FDR 1.664E-7, 3KCA = 3 ketocholanic acid, FDR 3.505E-7, 3a-H-12 KLCA = 3a-hydroxy-12 ketolithocholic acid, FDR 4.174E-6, CA = cholic acid, FDR 2.142E-7, DCA = deoxycholic acid, FDR 0.004, FDR = false discovery rate, ILCA = isolithocholic acid, FDR 1.596E-10, LCA = lithocholic acid, FDR 6.157E-10, PC = principal component, TCA = taurocholic acid, FDR 0.001.
Figure 3.
Figure 3.
Bacterial composition in age-matched critically ill and healthy children. Nonmetric multidimensional scaling (NMDS) plot of fecal microbial composition at genus level. High variability is seen in samples from critically ill children (red) compared with healthy controls (orange). PERmutational Multivariate ANalysis Of Variance (PERMANOVA) p = 0.0001, F = 3.314. Samples from healthy children were tightly clustered together, suggesting greater similarity in composition, while those from patients were scattered widely across the plot and statistically separated from the healthy profiles (PERMANOVA test: F = 9.78; p < 0.001).

References

    1. Modi SR, Collins JJ, Relman DA. Antibiotics and the gut microbiota. J Clin Invest 2014; 124:4212–4218
    1. Fouhy F, Guinane CM, Hussey S, et al. High-throughput sequencing reveals the incomplete, short-term recovery of infant gut microbiota following parenteral antibiotic treatment with ampicillin and gentamicin. Antimicrob Agents Chemother 2012; 56:5811–5820
    1. Yamada T, Shimizu K, Ogura H, et al. Rapid and sustained long-term decrease of fecal short-chain fatty acids in critically ill patients with systemic inflammatory response syndrome. J Parenter Enter Nutr 2015; 39:569–577
    1. Tedelind S, Westberg F, Kjerrulf M, et al. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: A study with relevance to inflammatory bowel disease. World J Gastroenterol 2007; 13:2826–2832
    1. Lankelma JM, van Vught LA, Belzer C, et al. Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation: A pilot study. Intensive Care Med 2017; 43:59–68
    1. Rogers MB, Firek B, Shi M, et al. Disruption of the microbiota across multiple body sites in critically ill children. Microbiome 2016; 4:66.
    1. Shimizu K, Ogura H, Goto M, et al. Altered gut flora and environment in patients with severe SIRS. J Trauma 2006; 60:126–133
    1. Zaborin A, Smith D, Garfield K, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio 2014; 5:e0136114
    1. Freedberg DE, Zhou MJ, Cohen ME, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med 2018; 44:1203–1211
    1. Correia GDS, Ng WK, Wijeyesekera A, et al. Metabolic profiling of children undergoing surgery for congenital heart disease. Crit Care Med 2015; 43:1467–1476
    1. Gaies MG, Gurney JG, Yen AH, et al. Vasoactive-inotropic score as a predictor of morbidity and mortality in infants after cardiopulmonary bypass. Pediatr Crit Care Med 2010; 11:234–238
    1. Gratton J, Phetcharaburanin J, Mullish BH, et al. Optimized sample handling strategy for metabolic profiling of human feces. Anal Chem 2016; 88:4661–4668
    1. Sarafian MH, Lewis MR, Pechlivanis A, et al. Bile acid profiling and quantification in biofluids using ultra-performance liquid chromatography tandem mass spectrometry. Anal Chem 2015; 87:9662–9670
    1. Wagner J, Coupland P, Browne HP, et al. Evaluation of PacBio sequencing for full-length bacterial 16S rRNA gene classification. BMC Microbiol 2016; 16:274.
    1. Oksanen J. Vegan: An introduction to ordination. Management 2008; 1:1–10
    1. Oksanen J, Blanchet FG, Friendly M, et al. Vegan: Community Ecology Package. R Package Vegan, Vers. 2.2-1, 2016. Available at: . Accessed May 23, 2019
    1. Wernovsky G, Wypij D, Jonas RA, et al. Postoperative course and hemodynamic profile after the arterial switch operation in neonates and infants. A comparison of low-flow cardiopulmonary bypass and circulatory arrest. Circulation 1995; 92:2226–2235
    1. Cummings JH. Short chain fatty acids in the human colon. Gut 1981; 22:763–779
    1. Bergman EN. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol Rev 1990; 70:567–590
    1. Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science 2012; 336:1262–1267
    1. Hatayama H, Iwashita J, Kuwajima A, et al. The short chain fatty acid, butyrate, stimulates MUC2 mucin production in the human colon cancer cell line, LS174T. Biochem Biophys Res Commun 2007; 356:599–603
    1. Ridlon JM, Kang DJ, Hylemon PB, et al. Bile acids and the gut microbiome. Curr Opin Gastroenterol 2014; 30:332–338
    1. Shindo K, Machida M, Fukumura M, et al. Omeprazole induces altered bile acid metabolism. Gut 1998; 42:266–271
    1. Zarrinpar A, Chaix A, Xu ZZ, et al. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nat Commun 2018; 9:2872.
    1. Toda T, Ohi K, Kudo T, et al. Ciprofloxacin suppresses Cyp3a in mouse liver by reducing lithocholic acid-producing intestinal flora. Drug Metab Pharmacokinet 2009; 24:201–208
    1. Zaborin A, Smith D, Garfield K, et al. Membership and behavior of ultra-low-diversity pathogen critical illness. MBio 2014; 5:1–14
    1. Kieser S, Sarker SA, Sakwinska O, et al. Bangladeshi children with acute diarrhoea show faecal microbiomes with increased Streptococcus abundance, irrespective of diarrhoea aetiology. Environ Microbiol 2018; 20:2256–2269
    1. Vincent J-L, Rello J, Marshall J, et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA 2009; 302:2323.
    1. McDonald D, Ackermann G, Khailova L, et al. Extreme dysbiosis of the microbiome in critical illness. mSphere 2016; 1:e0019916
    1. Livanos AE, Snider EJ, Whittier S, et al. Rapid gastrointestinal loss of Clostridial Clusters IV and XIVa in the ICU associates with an expansion of gut pathogens. PLoS One 2018; 13:e0200322.
    1. Scalabre A, Jobard E, Demède D, et al. Evolution of newborns’ urinary metabolomic profiles according to age and growth. J Proteome Res 2017; 16:3732–3740
    1. Lau CE, Siskos AP, Maitre L, et al. Determinants of the urinary and serum metabolome in children from six European populations. BMC Med 2018; 16:202.
    1. Bokulich NA, Chung J, Battaglia T, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016; 8:343ra82
    1. Wang C, Shoji H, Sato H, et al. Effects of oral administration of bifidobacterium breve on fecal lactic acid and short-chain fatty acids in low birth weight infants. J Pediatr Gastroenterol Nutr 2007; 44:252–257
    1. Nagpal R, Wang S, Ahmadi S, et al. Human-origin probiotic cocktail increases short-chain fatty acid production via modulation of mice and human gut microbiome. Sci Rep 2018; 8:12649.

Source: PubMed

3
Abonnieren