Profiling gut microbiota and bile acid metabolism in critically ill children

Iain Robert Louis Kean, Joseph Wagner, Anisha Wijeyesekera, Marcus De Goffau, Sarah Thurston, John A Clark, Deborah K White, Jenna Ridout, Shruti Agrawal, Riaz Kayani, Roddy O'Donnell, Padmanabhan Ramnarayan, Mark J Peters, Nigel Klein, Elaine Holmes, Julian Parkhill, Stephen Baker, Nazima Pathan, Iain Robert Louis Kean, Joseph Wagner, Anisha Wijeyesekera, Marcus De Goffau, Sarah Thurston, John A Clark, Deborah K White, Jenna Ridout, Shruti Agrawal, Riaz Kayani, Roddy O'Donnell, Padmanabhan Ramnarayan, Mark J Peters, Nigel Klein, Elaine Holmes, Julian Parkhill, Stephen Baker, Nazima Pathan

Abstract

Broad-spectrum antimicrobial use during the treatment of critical illness influences gastrointestinal fermentation endpoints, host immune response and metabolic activity including the conversion of primary to secondary bile acids. We previously observed reduced fermentation capacity in the faecal microbiota of critically ill children upon hospital admission. Here, we further explore the timecourse of the relationship between the microbiome and bile acid profile in faecal samples collected from critically ill children. The microbiome was assayed by sequencing of the 16S rRNA gene, and faecal water bile acids were measured by liquid chromatography mass spectrometry. In comparison to admission faecal samples, members of the Lachnospiraceae recovered during the late-acute phase (days 8-10) of hospitalisation. Patients with infections had a lower proportion of Lachnospiraceae in their gut microbiota than controls and patients with primary admitting diagnoses. Keystone species linked to ecological recovery were observed to decline with the length of PICU admission. These species were further suppressed in patients with systemic infection, respiratory failure, and undergoing surgery. Bile acid composition recovers quickly after intervention for critical illness which may be aided by the compositional shift in Lachnospiraceae. Our findings suggest gut microbiota recovery can be readily assessed via measurement of faecal bile acids.

Conflict of interest statement

The work in this study is supported by funding to NP from Action Medical Research, the Evelyn Trust and the European Society of Intensive Care Medicine. JP and SB received funding from The Wellcome Trust, and JP received funding from Next Gen Diagnostics Llc. The remaining authors have disclosed that they do not have any potential conflicts of interest.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
Primary and secondary bile acids (a) primary bile acids (Cholic acid (CA), Chenodeoxycholic acid (CDCA), Glycochenodeoxycholic acid (GCDCA), and Taurocholic acid (TCA)) in faecal water measured by LC–MS as a ratio of total measured bile acid. (b) secondary bile acids (Deoxycholic Acid (DCA), Lithocholic acid (LCA), Isolithocholic acid (ILCA), Ursodeoxycholic acid (UDCA), 3α-Hydroxy,12-Ketolithocholic acid (3a-H,12-KLCA), Taurohyocholic acid (THCA), and Glycoursodeoxycholic acid (GUDCA)) in faecal water measured by LC–MS as a ratio of total measured bile acid. A significant reduction to the relative concentration of total secondary bile acids versus the total measured bile acid pool was observed for patients sampled at day 1–3 and day 4–7 compared to the control. The median proportion of these secondary bile acids at days 8–10 was comparable to that of the aged-matched controls, but we observed a significant increase of patient bile acids at days 8–10 above the median for patients at day 1–3. (c) the ratio of CA to CDCA, two species of primary bile acids in the human gut. The control samples had a median CA:CDCA ratio of 0.9; the median CA:CDCA ratios on days 1–3, 4–7, an 8–10 in the faecal samples of the critically patients were 4.4, 4.4, and 5.9, respectively. (d) Ratio of primary bile acids to secondary bile acids. (e) ratio of CA to DCA in faecal water. The ratio of CA to DCA was elevated in across all patient timepoints compared to the control. Patient samples collected on days 8–10 had lower ratios of CA:DCA than patients at day 1–3. (f) ratio of CDCA to LCA and ILCA. CDCA:LCA ratios were increased compared to controls in patient samples from day 1–3 and day 4–7, returning to control levels at days 8–10. Conover-Iman significance ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05, p > 0.05 = not shown.
Figure 2
Figure 2
Bacterial diversity, and abundance of beneficial bacteria. (a) Shannon diversity of sampling groups, bars indicate 1st 2nd and 3rd quartiles. Due to the broad range of alpha-diversity at day 1–3, day 4–7 and days 8–10, only the median alpha diversity of samples collected at days 8–10 was significantly lower than that of the control group after correcting for multiple comparisons. (b) NMDS clustering of beta diversity of bacterial populations compared between sampling groups. PERMANOVA p-value = 0.001. Ellipses indicate 60% similarity. Conover-Iman significance ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05, p > 0.05 = not shown.
Figure 3
Figure 3
Relative proportion of bacteria identified by 16S rRNA gene sequencing. Bacterial 16S rRNA gene counts are represented as proportional abundance at Family level for each sample. The majority of analysed 16S rRNA gene counts are within 12k to 16k reads. The ten most abundant families in the control microbiomes were the Bacteroidaceae, Ruminococcaceae, Prevotellaceae, Lachnospiraceae, Tannerellaceae, Veillonellacea, Peptostreptococcaceae, Coriobacteriaceae, Rikenellaceae and Acidaminococcaceae. These ten families represent a median of 82.8% (IQR 12.7%) of the control faecal microbiota and a median of 45.3% (IQR 57%) of the patient faecal microbiota. The ten most abundant bacterial families present in the patient faecal microbiome, which were not highly represented in the control samples were the Enterococcaceae, Enterobacteriacea, Bifidobacteriaceae, Streptococcaceae, Staphylococcaceae, Corynebacteriaceae, Clostridiaceae, Lactobacillaceae, Christensenellaceae and the Sutterellaceae. These families represented a median of 7.9% (IQR 8.8%) of the control microbiome, and a median of 31.1% (IQR 64.8%) of the patient samples.
Figure 4
Figure 4
Relative abundance of recovery associated bacteria (RAB) and Lachnospiraceae. (a) Proportion of RAB present in microbiomes. Patient samples from day 1–3, day 4–7 and days 8–10 were significantly reduced from the median level of the control group. RAB at days 8–10 were also significantly reduced from the median RAB level at day 4–7. (b) Proportion of Lachnospiraceae present in microbiomes. No significant difference was observed between groups after controlling for multiple comparisons. Conover-Iman significance ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05, p > 0.05 = not shown.

References

    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. Milani C, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol. Mol. Biol. Rev. 2017;81:e00036-17. doi: 10.1128/MMBR.00036-17.
    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(1):1–16. doi: 10.1186/s13073-016-0294-z.
    1. Arrieta MC, Stiemsma LT, Amenyogbe N, Brown E, Finlay B. The intestinal microbiome in early life: Health and disease. Front. Immunol. 2014;5:427. doi: 10.3389/fimmu.2014.00427.
    1. Stewart CJ, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562:583–588. doi: 10.1038/s41586-018-0617-x.
    1. Ruohtula T, et al. Maturation of gut microbiota and circulating regulatory T cells and development of IgE sensitization in early life. Front. Immunol. 2019;10:2494. doi: 10.3389/fimmu.2019.02494.
    1. Heida FH, et al. A necrotizing enterocolitis-associated gut microbiota is present in the meconium: results of a prospective study. Clin. Infect. Dis. 2016;62:863–870. doi: 10.1093/cid/ciw016.
    1. Rinninella E, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019;7:14. doi: 10.3390/microorganisms7010014.
    1. Dickson RP. The microbiome and critical illness. Lancet Respir. Med. 2016;4:59–72. doi: 10.1016/S2213-2600(15)00427-0.
    1. Wijeyesekera A, et al. Multi-compartment profiling of bacterial and host metabolites identifies intestinal dysbiosis and its functional consequences in the critically Ill child. Crit. Care Med. 2019;47:e727–e734. doi: 10.1097/CCM.0000000000003841.
    1. Rogers MB, et al. Disruption of the microbiota across multiple body sites in critically ill children. Microbiome. 2016;4:66. doi: 10.1186/s40168-016-0211-0.
    1. Chng KR, et al. Metagenome-wide association analysis identifies microbial determinants of post-antibiotic ecological recovery in the gut. Nat. Ecol. Evol. 2020;4(9):1256–1267. doi: 10.1038/s41559-020-1236-0.
    1. Moron R, et al. The importance of the microbiome in critically ill patients: Role of nutrition. Nutrients. 2019;11(12):3002. doi: 10.3390/nu11123002.
    1. Moreno-Indias I, et al. Intermittent hypoxia alters gut microbiota diversity in a mouse model of sleep apnoea. Eur. Respir. J. 2015;45:1055–1065. doi: 10.1183/09031936.00184314.
    1. Looft T, et al. In-feed antibiotic effects on the swine intestinal microbiome. Proc. Natl. Acad. Sci. 2012;109(5):1691–1696. doi: 10.1073/pnas.1120238109.
    1. Schubert K, Olde Damink SWM, von Bergen M, Schaap FG. Interactions between bile salts, gut microbiota, and hepatic innate immunity. Immunol. Rev. 2017;279:23–35. doi: 10.1111/imr.12579.
    1. Urdaneta V, Casadesús J. Interactions between bacteria and bile salts in the gastrointestinal and hepatobiliary tracts. Front. Med. 2017;4:163. doi: 10.3389/fmed.2017.00163.
    1. Heinken A, et al. Systematic assessment of secondary bile acid metabolism in gut microbes reveals distinct metabolic capabilities in inflammatory bowel disease. Microbiome. 2019;7:75. doi: 10.1186/s40168-019-0689-3.
    1. Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 2006;47:241–259. doi: 10.1194/jlr.R500013-JLR200.
    1. Kang JD, et al. Bile acid 7α-dehydroxylating gut bacteria secrete antibiotics that inhibit clostridium difficile: role of secondary bile acids. Cell Chem. Biol. 2019;26:27–34.e4. doi: 10.1016/j.chembiol.2018.10.003.
    1. Solbach P, et al. BaiCD gene cluster abundance is negatively correlated with Clostridium difficile infection. PLoS ONE. 2018;13:e0196977. doi: 10.1371/journal.pone.0196977.
    1. Staley C, Weingarden AR, Khoruts A, Sadowsky MJ. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl. Microbiol. Biotechnol. 2017;101:47–64. doi: 10.1007/s00253-016-8006-6.
    1. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr. Opin. Gastroenterol. 2014;30:332–338. doi: 10.1097/MOG.0000000000000057.
    1. Ridlon JM, Wolf PG, Gaskins HR. Taurocholic acid metabolism by gut microbes and colon cancer. Gut Microbes. 2016;7:201–215. doi: 10.1080/19490976.2016.1150414.
    1. Ma H, Patti ME, Endocrinologist A. Bile acids, obesity, and the metabolic syndrome. Best Pract. Res. Clin. Gastroenterol. 2014;28:573–583. doi: 10.1016/j.bpg.2014.07.004.
    1. Festi D, et al. Gut microbiota and metabolic syndrome. World J. Gastroenterol. 2014;20:16079–16094. doi: 10.3748/wjg.v20.i43.16079.
    1. Reddy BS, Narasawa T, Weisburger JH, Wynder EL. Promoting effect of sodium deoxycholate on colon adenocarcinomas in germfree Rats2. JNCI J. Natl. Cancer Inst. 1976;56:441–442. doi: 10.1093/jnci/56.2.441.
    1. Reddy BS, Watanabe K, Weisburger JH, Wynder EL. Promoting effect of bile acids in colon carcinogenesis in germ-free and conventional F344 rats. Cancer Res. 1977;37:3238–3242.
    1. Attene-Ramos MS, Wagner ED, Gaskins HR, Plewa MJ. Hydrogen sulfide induces direct radical-associated DNA damage. Mol. Cancer Res. 2007;5:455–459. doi: 10.1158/1541-7786.MCR-06-0439.
    1. Devkota S, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature. 2012;487:104–108. doi: 10.1038/nature11225.
    1. Shetty SA, et al. Reclassification of Eubacterium hallii as Anaerobutyricum hallii gen. nov., comb. Nov., and description of Anaerobutyricum soehngenii sp. Nov., a butyrate and propionate-producing bacterium from infant faeces. Int. J. Syst. Evol. Microbiol. 2018;68:3741–3746. doi: 10.1099/ijsem.0.003041.
    1. Togo, A. H. et al. Description of Mediterraneibacter massiliensis, gen. nov., sp. nov., a new genus isolated from the gut microbiota of an obese patient and reclassification of Ruminococcus faecis, Ruminococcus lactaris, Ruminococcus torques, Ruminococcus gnavus and Clostridium glycyrrhizinilyticum as Mediterraneibacter faecis comb. nov., Mediterraneibacter lactaris comb. nov., Mediterraneibacter torques comb. nov., Mediterraneibacter gnavus comb. nov. and Mediterraneibacter glycyrrhizinilyticus comb. nov. Antonie Van Leeuwenhoek111, 2107–2128 (2018).
    1. Hirano S, Nakama R, Tamaki M, Masuda N, Oda H. Isolation and characterization of thirteen intestinal microorganisms capable of 7α-dehydroxylating bile acids. Appl. Environ. Microbiol. 1981;41:737–745. doi: 10.1128/aem.41.3.737-745.1981.
    1. Berr F, Kullak-Ublick GA, Paumgartner G, Monzing W, Hylemon PB. 7α-Dehydroxylating bacteria enhance deoxycholic acid input and cholesterol saturation of bile in patients with gallstones. Gastroenterology. 1996;111:1611–1620. doi: 10.1016/S0016-5085(96)70024-0.
    1. Chiang JYL. Bile acids: regulation of synthesis. J. Lipid Res. 2009;50:1955–1966. doi: 10.1194/jlr.R900010-JLR200.
    1. Song Z, et al. Taxonomic profiling and populational patterns of bacterial bile salt hydrolase (BSH) genes based on worldwide human gut microbiome. Microbiome. 2019;7:9. doi: 10.1186/s40168-019-0628-3.
    1. Stellwag EJ, Hylemon PB. 7alpha-Dehydroxylation of cholic acid and chenodeoxycholic acid by Clostridium leptum. J. Lipid Res. 1979;20:325–333. doi: 10.1016/S0022-2275(20)40615-7.
    1. Batta AK, et al. Side chain conjugation prevents bacterial 7-dehydroxylation of bile acids. J. Biol. Chem. 1990;265:10925–10928. doi: 10.1016/S0021-9258(19)38535-7.
    1. Van Eldere J, Celis P, De Pauw G, Lesaffre E, Eyssen H. Tauroconjugation of cholic acid stimulates 7 alpha-dehydroxylation by fecal bacteria. Appl. Environ. Microbiol. 1996;62:656–661. doi: 10.1128/aem.62.2.656-661.1996.
    1. Vital M, Rud T, Rath S, Pieper DH, Schlüter D. Diversity of bacteria exhibiting bile acid-inducible 7α-dehydroxylation genes in the human gut. Comput. Struct. Biotechnol. J. 2019;17:1016–1019. doi: 10.1016/j.csbj.2019.07.012.
    1. Gibbons SM. Keystone taxa indispensable for microbiome recovery. Nat. Microbiol. 2020;5:1067–1068. doi: 10.1038/s41564-020-0783-0.
    1. Paine RT. A conversation on refining the concept of keystone species. Conserv. Biol. 1995;9:962–964. doi: 10.1046/j.1523-1739.1995.09040962.x.
    1. Begley M, Hill C, Gahan CGM. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 2006;72:1729–1738. doi: 10.1128/AEM.72.3.1729-1738.2006.
    1. Marion S, et al. In vitro and in vivo characterization of Clostridium scindens bile acid transformations. Gut Microbes. 2018;10:481–503. doi: 10.1080/19490976.2018.1549420.
    1. Duncan SH, Louis P, Flint HJ. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl. Environ. Microbiol. 2004;70:5810–5817. doi: 10.1128/AEM.70.10.5810-5817.2004.
    1. PICANet_2019_Annual_Report_Tables_and_Figures_v1.0.pdf.
    1. Neu J, Rushing J. Cesarean versus vaginal delivery: long-term infant outcomes and the hygiene hypothesis. Clin. Perinatol. 2011;38:321–331. doi: 10.1016/j.clp.2011.03.008.
    1. Biasucci G, Benenati B, Morelli L, Bessi E, Boehm G. Cesarean delivery may affect the early biodiversity of intestinal bacteria. J. Nutr. 2008 doi: 10.1093/jn/138.9.1796s.
    1. Dominguez-Bello MG, et al. Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat. Med. 2016;22:250–253. doi: 10.1038/nm.4039.
    1. Yassour M, et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 2016;8:343ra81. doi: 10.1126/scitranslmed.aad0917.
    1. Sarafian MH, et al. Bile acid profiling and quantification in biofluids using ultra-performance liquid chromatography tandem mass spectrometry. Anal. Chem. 2015;87:9662–9670. doi: 10.1021/acs.analchem.5b01556.
    1. Kakiyama G, et al. A simple and accurate HPLC method for fecal bile acid profile in healthy and cirrhotic subjects: Validation by GC-MS and LC-MS. J. Lipid Res. 2014;55:978–990. doi: 10.1194/jlr.D047506.
    1. Strathmann FG, Hoofnagle AN. Current and future applications of mass spectrometry to the clinical laboratory. Am. J. Clin. Pathol. 2011;136:609–616. doi: 10.1309/AJCPW0TA8OBBNGCK.
    1. Seger C, Salzmann L. After another decade: LC–MS/MS became routine in clinical diagnostics. Clin. Biochem. 2020;82:2–11. doi: 10.1016/j.clinbiochem.2020.03.004.
    1. Bokulich NA, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016;8:343ra82. doi: 10.1126/scitranslmed.aad7121.
    1. Wang C, 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 doi: 10.1097/01.mpg.0000252184.89922.5f.
    1. Suez J, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174:1406–1423.e16. doi: 10.1016/j.cell.2018.08.047.
    1. Mallick H, et al. Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences. Nat. Commun. 2019;10:1–11. doi: 10.1038/s41467-019-10927-1.
    1. Larsen PE, Dai Y. Metabolome of human gut microbiome is predictive of host dysbiosis. GigaScience. 2015 doi: 10.1186/s13742-015-0084-3.
    1. Schloss PD, et al. Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009;75:7537–7541. doi: 10.1128/AEM.01541-09.
    1. Gratton J, et al. Optimized Sample Handling Strategy for Metabolic Profiling of Human Feces. Anal. Chem. 2016;88:4661–4668. doi: 10.1021/acs.analchem.5b04159.
    1. Shockcor, J., Crowe, H., Yu, K. & Shion, H. Analysis of Intact Lipids from Biologics Matrices by UPLC / Ion Mobility TOF-MS. Waters Corp. Milford MA USA 2–4 (2011).
    1. R Core Team. R software: Version 4.0.2. R Found. Stat. Comput. (2020).
    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. Conover WJ, Iman RL. Rank transformations as a bridge between parametric and nonparametric statistics. Am. Stat. 1981;35:124.
    1. Dinno, A. conover.test: Conover-Iman Test of Multiple Comparisons Using Rank Sums. (2017).
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Methodol. 1995;57:289–300.
    1. Wickham H. ggplot2 Elegant Graphics for Data Analysis (Use R!) Springer. 2016 doi: 10.1007/978-0-387-98141-3.
    1. Wickham, H. R: Package ‘reshape2’. CRAN (2017).
    1. Guevarra, E. nutriverse/zscorer: zscorer v0.3.1. (Zenodo, 2019). 10.5281/zenodo.3510075.
    1. Chou JH, Roumiantsev S, Singh R. PediTools electronic growth chart calculators: applications in clinical care, research, and quality improvement. J. Med. Internet Res. 2020;22:e16204. doi: 10.2196/16204.
    1. Oksanen, J. et al. vegan: Community Ecology Package. R package version 2.5–2. Cran R (2019).
    1. 11. Correlation and regression | The BMJ. The BMJ | The BMJ: leading general medical journal. Research. Education. Comment (2020).

Source: PubMed

3
Tilaa