Skip to main content
Top

27-10-2016 | Microbiome | Article

Fecal metabolomics in pediatric spondyloarthritis implicate decreased metabolic diversity and altered tryptophan metabolism as pathogenic factors

Authors: M L Stoll, R Kumar, E J Lefkowitz, R Q Cron, C D Morrow, S Barnes

Abstract

We have previously shown alterations in the composition of the gut microbiota in children with enthesitis-related arthritis (ERA). To explore the mechanisms by which an altered microbiota might predispose to arthritis, we performed metabolomic profiling of fecal samples of children with ERA. Fecal samples were collected from two cohorts of children with ERA and healthy control subjects. Nano-liquid chromatography—mass spectroscopy (LC-MS) was performed on the fecal water homogenates with identification based upon mass: charge ratios. Sequencing of the 16S ribosomal DNA (rDNA) on the same stool specimens was performed. In both sets of subjects, patients demonstrated lower diversity of ions and under-representation of multiple metabolic pathways, including the tryptophan metabolism pathway. For example, in the first cohort, out of 1500 negatively charged ions, 154 were lower in ERA patients, compared with only one that was higher. Imputed functional annotation of the 16S ribosomal DNA sequence data demonstrated significantly fewer microbial genes associated with metabolic processes in the patients compared with the controls (77 million versus 58 million, P=0.050). Diminished metabolic diversity and alterations in the tryptophan metabolism pathway may be a feature of ERA.

Genes Immun 2016;17:400–405. doi:10.1038/gene.2016.38

Interest in the composition of the gut microbiota in subjects with spondyloarthritis (SpA), like enthesitis-related arthritis (ERA)/juvenile idiopathic arthritis (JIA), has been accumulating.1 We and others have identified taxonomic differences in fecal bacteria between pediatric or adult SpA subjects and healthy controls.2, 3, 4 However, these studies have not provided a mechanism whereby dysbiosis can result in arthritis. One possible mechanism is through alterations in metabolic pathways. The metabolic capacity of bacteria is an under-appreciated aspect of the human microbiome. In total, bacteria contain over 3 million genes, 100 times the human host;5 they perform a variety of metabolic functions including metabolism of dietary components, drug detoxification and synthesis of vitamins and essential amino acids.5 Bacteria are not created equal in their capacity to perform these functions, and thus a particular microbiome may be more or less effective than another at carrying out certain activities. To evaluate the functional potential of the microbiome, fecal water metabolomics on children with ERA and controls was performed. The aim of metabolomics is to conduct a comprehensive analysis on the identities of the low molecular weight ions (in our facility, <1000 Da) present in a sample, so as to obtain insight into function.6Metabolomics of fecal water, the supernatant obtained following high-speed centrifugation of feces, can discriminate between inflammatory bowel disease (IBD) patients and healthy individuals, with some studies showing elevated levels of amino acids and decreased short chain fatty acids such as butyrate.7, 8 Our intent was to identify mechanisms by which the microbiota might predispose or contribute to arthritis in children with ERA.

Literature
  1. Burns, C. M. & Wortmann, R. L. Gout therapeutics: new drugs for an old disease. Lancet 377, 165–177 (2011).
  2. Richette, P. & Bardin, T. Gout. Lancet 375, 318–328 (2010).
  3. Zhu, Y., Pandya, B. J. & Choi, H. K. Comorbidities of gout and hyperuricemia in the US general population: NHANES 2007–2008. Am J Med 125, 679–687 (2012).
  4. Kuo, C. F., Grainge, M. J., Mallen, C., Zhang, W. & Doherty, M. Rising burden of gout in the UK but continuing suboptimal management: a nationwide population study. Ann Rheum Dis 74, 661–667 (2014).
  5. Guo, M. et al. Polymorphism of rs7688672 and rs10033237 in cGKII/PRKG2 and gout susceptibility of Han population in northern China. Gene 562, 50–54 (2015).
  6. Krishnan, E. et al. Serum urate and incidence of kidney disease among veterans with gout. J Rheumatol 40, 1166–1172 (2013).
  7. Lee, S. et al. Uric Acid Level Is Not An Independent Predictor Of Cardiovascular Diseases In Gout Patients With Treatment; Long-Term Follow-Up Data In Single Tertiary Center In South Korea. Arthritis and rheumatism 65, S856–S856 (2013).
  8. McQueen, F. M. Gout in 2013. Imaging, genetics and therapy: gout research continues apace. Nat Rev Rheumatol 10, 67–69 (2014).
  9. Chang, W. C. Dietary intake and the risk of hyperuricemia, gout and chronic kidney disease in elderly Taiwanese men. Aging Male 14, 195–202 (2011).
  10. Sorensen, L. B. & Levinson, D. J. Origin and extrarenal elimination of uric acid in man. Nephron 14, 7–20 (1975).
  11. Nicholson, J. K. et al. Host-gut microbiota metabolic interactions. Science 336, 1262–1267 (2012).
  12. Kau, A. L., Ahern, P. P., Griffin, N. W., Goodman, A. L. & Gordon, J. I. Human nutrition, the gut microbiome and the immune system. Nature 474, 327–336 (2011).
  13. Jumpertz, R. et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr 94, 58–65 (2011).
  14. Gordon, J. I., Dewey, K. G., Mills, D. A. & Medzhitov, R. M. The Human Gut Microbiota and Undernutrition. Sci Transl Med 4, (2012).
  15. Xu, J. & Gordon, J. I. Honor thy symbionts. Proc Natl Acad Sci USA 100, 10452–10459 (2003).
  16. Crane, J. K., Naeher, T. M., Broome, J. E. & Boedeker, E. C. Role of host xanthine oxidase in infection due to enteropathogenic and Shiga-toxigenic Escherichia coli. Infect Immun 81, 1129–1139 (2013).
  17. Sathisha, K. R. et al. Synthesis and xanthine oxidase inhibitory activity of 7-methyl-2-(phenoxymethyl)-5H-[1,3,4]thiadiazolo[3,2-a] pyrimidin-5-one derivatives. Bioorg Med Chem 19, 211–220 (2011).
  18. Hsieh, C. Y., Lin, H. J., Chen, C. H., Lai, E. C. & Yang, Y. H. Chronic kidney disease and stroke. Lancet Neurol 13, 1071 (2014).
  19. Hosomi, A., Nakanishi, T., Fujita, T. & Tamai, I. Extra-renal elimination of uric acid via intestinal efflux transporter BCRP/ABCG2. PloS One 7, e30456 (2012).
  20. Nielsen, H. B. et al. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat Biotechnol 32, 822–828 (2014).
  21. Wei, B. et al. Molecular cloning of a Bacteroides caccae TonB-linked
  22. Ulsemer, P. et al. Safety and tolerance of Bacteroides xylanisolvens DSM 23964 in healthy adults. Benef Microbes 3, 99–111 (2012).
  23. Ulsemer, P., Toutounian, K., Schmidt, J., Karsten, U. & Goletz, S. Preliminary safety evaluation of a new Bacteroides xylanisolvens isolate. Appl Environ Microbiol 78, 528–535 (2012).
  24. Eckburg, P. B. et al. Diversity of the human intestinal microbial flora. Science 308, 1635–1638 (2005).
  25. Li, G., Yao, W. & Jiang, H. Short-chain fatty acids enhance adipocyte differentiation in the stromal vascular fraction of porcine adipose tissue. J Nutr 144, 1887–1895 (2014).
  26. Qin, N. et al. Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 59–64 (2014).
  27. Qin, J. J. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60 (2012).
  28. Karlsson, F. H. et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 498, 99–103 (2013).
  29. Griffiths, M. The mechanism of the diabetogenic action of uric acid. J Biol Chem 184, 289–298 (1950).
  30. Tsouli, S. G., Liberopoulos, E. N., Mikhailidis, D. P., Athyros, V. G. & Elisaf, M. S. Elevated serum uric acid levels in metabolic syndrome: an active component or an innocent bystander? Metabolism 55, 1293–1301 (2006).
  31. Facchini, F., Chen, Y. D., Hollenbeck, C. B. & Reaven, G. M. Relationship between resistance to insulin-mediated glucose uptake, urinary uric acid clearance, and plasma uric acid concentration. Jama 266, 3008–3011 (1991).
  32. Li, J. et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol 32, 834–841 (2014).
  33. Matsuo, H. et al. ABCG2 dysfunction causes hyperuricemia due to both renal urate underexcretion and renal urate overload. Sci Rep 4, 3375 (2014).
  34. Caporaso, J. G. et al. PyNAST: a flexible tool for aligning sequences to a template alignment. Bioinformatics 26, 266–267 (2010).
  35. Claesson, M. J. et al. Comparative Analysis of Pyrosequencing and a Phylogenetic Microarray for Exploring Microbial Community Structures in the Human Distal Intestine. PloS One 4, e6669 (2009).
  36. Peng, Y., Leung, H. C. M., Yiu, S. M. & Chin, F. Y. L. IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth. Bioinformatics 28, 1420–1428 (2012).
  37. Zhu, W. H., Lomsadze, A. & Borodovsky, M. Ab initio gene identification in metagenomic sequences. Nucleic Acids Research 38, e132 (2010).
  38. Li, W. Z. & Godzik, A. Cd-hit: a fast program for clustering and comparing large sets of protein or nucleotide sequences. Bioinformatics 22, 1658–1659 (2006).
  39. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biology 10, R25 (2009).
  40. McArdle, B. H. & Anderson, M. J. Fitting multivariate models to community data: A comment on distance-based redundancy analysis. Ecology 82, 290–297 (2001).
  41. Zapala, M. A. & Schork, N. J. Multivariate regression analysis of distance matrices for testing associations between gene expression patterns and related variables. Proc Natl Acad Sci USA 103, 19430–19435 (2006).
  42. Robin, X. et al. pROC: an open-source package for R and S plus to analyze and compare ROC curves. Bmc Bioinformatics 12, 77 (2011).