Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile (original) (raw)

Accession codes

Primary accessions

Sequence Read Archive

Data deposits

Study sequence data are deposited in the National Center for Biotechnology Information Sequence Read Archive under accession number SRP045811.

Change history

A minor change was made to the author list.

References

  1. Buffie, C. G. & Pamer, E. G. Microbiota-mediated colonization resistance against intestinal pathogens. Nature Rev. Immunol. 13, 790–801 (2013)
    Article CAS Google Scholar
  2. Buffie, C. G. et al. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium _difficile_-induced colitis. Infect. Immun. 80, 62–73 (2012)
    Article CAS Google Scholar
  3. Rupnik, M., Wilcox, M. H. & Gerding, D. N. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nature Rev. Microbiol. 7, 526–536 (2009)
    Article CAS Google Scholar
  4. van Nood, E. et al. Duodenal infusion of donor feces for recurrent Clostridium difficile . N. Engl. J. Med. 368, 407–415 (2013)
    Article CAS Google Scholar
  5. Cimermancic, P. et al. Insights into secondary metabolism from a global analysis of prokaryotic biosynthetic gene clusters. Cell 158, 412–421 (2014)
    Article CAS Google Scholar
  6. Chang, J. Y. et al. Decreased diversity of the fecal microbiome in recurrent Clostridium _difficile_-associated diarrhea. J. Infect. Dis. 197, 435–438 (2008)
    Article Google Scholar
  7. Lozupone, C. & Knight, R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl. Environ. Microbiol. 71, 8228–8235 (2005)
    Article CAS Google Scholar
  8. Taur, Y. et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis. 55, 905–914 (2012)
    Article CAS Google Scholar
  9. Kinnebrew, M. A. et al. Early Clostridium difficile infection during allogeneic hematopoietic stem cell transplantation. PLoS ONE 9, e90158 (2014)
    Article ADS Google Scholar
  10. Stein, R. R. et al. Ecological modeling from time-series inference: insight into dynamics and stability of intestinal microbiota. PLOS Comput. Biol. 9, e1003388 (2013)
    Article Google Scholar
  11. Wilson, K. H. Efficiency of various bile salt preparations for stimulation of Clostridium difficile spore germination. J. Clin. Microbiol. 18, 1017–1019 (1983)
    CAS PubMed PubMed Central Google Scholar
  12. Sorg, J. A. & Sonenshein, A. L. Bile salts and glycine as cogerminants for Clostridium difficile spores. J. Bacteriol. 190, 2505–2512 (2008)
    Article CAS Google Scholar
  13. Kang, D. J., Ridlon, J. M., Moore, D. R., Barnes, S. & Hylemon, P. B. Clostridium scindens baiCD and baiH genes encode stereo-specific 7α/7β-hydroxy-3-oxo-Δ4-cholenoic acid oxidoreductases. Biochim. Biophys. Acta 1781, 16–25 (2008)
    Article CAS Google Scholar
  14. Ridlon, J. M., Kang, D. J. & Hylemon, P. B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 47, 241–259 (2006)
    Article CAS Google Scholar
  15. Langille, M. G. et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nature Biotechnol. 31, 814–821 (2013)
    Article CAS Google Scholar
  16. Out, C., Groen, A. K. & Brufau, G. Bile acid sequestrants: more than simple resins. Curr. Opin. Lipidol. 23, 43–55 (2012)
    Article CAS Google Scholar
  17. Collins, M. D. et al. The phylogeny of the genus Clostridium: proposal of five new genera and eleven new species combinations. Int. J. Syst. Bacteriol. 44, 812–826 (1994)
    Article CAS Google Scholar
  18. Yutin, N. & Galperin, M. Y. A genomic update on clostridial phylogeny: Gram-negative spore formers and other misplaced clostridia. Environ. Microbiol. 15, 2631–2641 (2013)
    CAS PubMed PubMed Central Google Scholar
  19. Kitahara, M., Takamine, F., Imamura, T. & Benno, Y. Assignment of Eubacterium sp. VPI 12708 and related strains with high bile acid 7α-dehydroxylating activity to Clostridium scindens and proposal of Clostridium hylemonae sp. nov., isolated from human faeces. Int. J. Syst. Evol. Microbiol. 50, 971–978 (2000)
    Article CAS Google Scholar
  20. Wells, E. & Hylemon, B. Identification and characterization of a bile acid 7α-dehydroxylation operon in Clostridium sp. strain TO-931, a highly active 7α-dehydroxylating strain isolated from human feces. Appl. Environ. Microbiol. 66, 1107–1113 (2000)
    Article CAS Google Scholar
  21. Weingarden, A. R. et al. Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficile infection. Am. J. Physiol. Gastrointest. Liver Physiol. 306, G310–G319 (2014)
    Article CAS Google Scholar
  22. Bernstein, H., Bernstein, C., Payne, C. M., Dvorakova, K. & Garewal, H. Bile acids as carcinogens in human gastrointestinal cancers. Mutat. Res. 589, 47–65 (2005)
    Article CAS Google Scholar
  23. Ng, K. M. et al. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature 502, 96–99 (2013)
    Article ADS CAS Google Scholar
  24. Jarchum, I., Liu, M., Shi, C., Equinda, M. & Pamer, E. G. Critical role for MyD88-mediated neutrophil recruitment during Clostridium difficile colitis. Infect. Immun. 80, 2989–2996 (2012)
    Article CAS Google Scholar
  25. Jarchum, I., Liu, M., Lipuma, L. & Pamer, E. G. Toll-like receptor 5 stimulation protects mice from acute Clostridium difficile colitis. Infect. Immun. 79, 1498–1503 (2011)
    Article CAS Google Scholar
  26. Rea, M. C. et al. Thuricin CD, a posttranslationally modified bacteriocin with a narrow spectrum of activity against Clostridium difficile . Proc. Natl Acad. Sci. USA 107, 9352–9357 (2010)
    Article ADS CAS Google Scholar
  27. Chen, X. et al. A mouse model of _Clostridium difficile_-associated disease. Gastroenterology 135, 1984–1992 (2008)
    Article Google Scholar
  28. Giel, J. L., Sorg, J. A., Sonenshein, A. L. & Zhu, J. Metabolism of bile salts in mice influences spore germination in Clostridium difficile . PLoS ONE 5, e8740 (2010)
    Article ADS Google Scholar
  29. Ubeda, C. et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J. Clin. Invest. 120, 4332–4341 (2010)
    Article CAS Google Scholar
  30. Wells, J. E., Williams, K. B., Whitehead, T. R., Heuman, D. M. & Hylemon, P. B. Development and application of a polymerase chain reaction assay for the detection and enumeration of bile acid 7α-dehydroxylating bacteria in human feces. Clin. Chim. Acta 331, 127–134 (2003)
    Article CAS Google Scholar
  31. Caporaso, J. G. et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 6, 1621–1624 (2012)
    Article CAS Google Scholar
  32. Ubeda, C. et al. Intestinal microbiota containing Barnesiella cures vancomycin-resistant Enterococcus faecium colonization. Infect. Immun. 81, 965–973 (2013)
    Article CAS Google Scholar
  33. Schloss, P. D. et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 75, 7537–7541 (2009)
    Article CAS Google Scholar
  34. Edgar, R. C., Haas, B. J., Clemente, J. C., Quince, C. & Knight, R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27, 2194–2200 (2011)
    Article CAS Google Scholar
  35. Sheneman, L., Evans, J. & Foster, J. A. Clearcut: a fast implementation of relaxed neighbor joining. Bioinformatics 22, 2823–2824 (2006)
    Article CAS Google Scholar
  36. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nature Methods 7, 335–336 (2010)
    Article CAS Google Scholar
  37. Hall, B. G. Building phylogenetic trees from molecular data with MEGA. Mol. Biol. Evol. 30, 1229–1235 (2013)
    Article CAS Google Scholar
  38. Human Microbiome Project Consortium Structure. Function and diversity of the healthy human microbiome. Nature 486, 207–214 (2012)
  39. Zhao, Y., Tang, H. & Ye, Y. RAPSearch2: a fast and memory-efficient protein similarity search tool for next-generation sequencing data. Bioinformatics 28, 125–126 (2012)
    Article CAS Google Scholar
  40. Cohen, J. Statistical Power Analysis for the Behavioral Sciences (Routledge, 1988)
    MATH Google Scholar

Download references

Acknowledgements

E.G.P. received funding from US National Institutes of Health (NIH) grants RO1 AI42135 and AI95706, and from the Tow Foundation. J.B.X. received funding from the NIH Office of the Director (DP2OD008440), NCI (U54 CA148967), and from a seed grant from the Lucille Castori Center for Microbes, Inflammation, and Cancer. C.G.B. was supported by a Medical Scientist Training Program grant from the National Institute of General Medical Sciences of the NIH (award number T32GM07739, awarded to the Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program).

Author information

Authors and Affiliations

  1. Department of Medicine, Infectious Diseases Service, Memorial Sloan Kettering Cancer Center, New York, 10065, New York, USA
    Charlie G. Buffie, Peter T. McKenney, Melissa Kinnebrew, Ying Taur & Eric G. Pamer
  2. Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, 10065, New York, USA
    Charlie G. Buffie, Peter T. McKenney, Lilan Ling, Asia Gobourne, Daniel No, Melissa Kinnebrew, Eric Littmann, Ying Taur, Nora C. Toussaint, Joao B. Xavier & Eric G. Pamer
  3. Computational Biology Program, Sloan-Kettering Institute, New York, 10065, New York, USA
    Vanni Bucci, Richard R. Stein, Chris Sander, Nora C. Toussaint & Joao B. Xavier
  4. Department of Biology, University of Massachusetts Dartmouth, North Dartmouth, 02747, Massachusetts, USA
    Vanni Bucci
  5. Donald B. and Catherine C. Marron Cancer Metabolism Center, Sloan-Kettering Institute, New York, 10065, New York, USA
    Hui Liu & Justin R. Cross
  6. Genomics Core Laboratory, Sloan-Kettering Institute, New York, 10065, New York, USA
    Agnes Viale
  7. Department of Medicine, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, 10065, New York, USA
    Marcel R. M. van den Brink & Robert R. Jenq
  8. Immunology Program, Sloan-Kettering Institute, New York, 10065, New York, USA
    Marcel R. M. van den Brink & Eric G. Pamer

Authors

  1. Charlie G. Buffie
    You can also search for this author inPubMed Google Scholar
  2. Vanni Bucci
    You can also search for this author inPubMed Google Scholar
  3. Richard R. Stein
    You can also search for this author inPubMed Google Scholar
  4. Peter T. McKenney
    You can also search for this author inPubMed Google Scholar
  5. Lilan Ling
    You can also search for this author inPubMed Google Scholar
  6. Asia Gobourne
    You can also search for this author inPubMed Google Scholar
  7. Daniel No
    You can also search for this author inPubMed Google Scholar
  8. Hui Liu
    You can also search for this author inPubMed Google Scholar
  9. Melissa Kinnebrew
    You can also search for this author inPubMed Google Scholar
  10. Agnes Viale
    You can also search for this author inPubMed Google Scholar
  11. Eric Littmann
    You can also search for this author inPubMed Google Scholar
  12. Marcel R. M. van den Brink
    You can also search for this author inPubMed Google Scholar
  13. Robert R. Jenq
    You can also search for this author inPubMed Google Scholar
  14. Ying Taur
    You can also search for this author inPubMed Google Scholar
  15. Chris Sander
    You can also search for this author inPubMed Google Scholar
  16. Justin R. Cross
    You can also search for this author inPubMed Google Scholar
  17. Nora C. Toussaint
    You can also search for this author inPubMed Google Scholar
  18. Joao B. Xavier
    You can also search for this author inPubMed Google Scholar
  19. Eric G. Pamer
    You can also search for this author inPubMed Google Scholar

Contributions

C.G.B. and E.G.P. designed the experiments and wrote the manuscript with input from co-authors. C.G.B. performed animal experiments and most analyses. V.B., R.R.S., J.B.X., C.S. and C.G.B. performed microbiota time-series inference modelling and analysis. P.T.M. and C.G.B designed and performed ex vivo experiments. L.L., A.G., A.V. D.N. and M.K. performed 16S amplicon quantification and multiparallel sequencing (454, MiSeq) and contributed to data analysis. M.R.M.v.d.B., R.R.J., Y.T., E.L., C.G.B. and E.G.P. assessed clinical parameters and supervised patient cohort analysis. N.C.T. and C.G.B. performed metagenomic shotgun sequencing analysis. J.R.C. and H.L. developed the metabolomics analysis platform and performed quantification of bile acid species.

Corresponding author

Correspondence toEric G. Pamer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Dynamics of intestinal microbiota structure and C. difficile susceptibility after antibiotic exposure.

a, Strategy for determining C. difficile susceptibility duration post-antibiotic exposure (n = 3 separately-housed mouse colonies per antibiotic arm) and relating infection resistance to microbiota structure. b, Correlation of C. difficile c.f.u. and toxin in intestinal content following infection. c, Intestinal bacterial density of animals before and after antibiotic exposure. d, Relative abundance of bacterial OTUs (≥97% sequence similarity, >0.01% relative abundance) sorted by class (red) and corresponding C. difficile susceptibility (blue) among antibiotic-exposed mice (n = 68) allowed to recover for variable time intervals prior to C. difficile infection challenge. Centre values (mean), error bars (s.e.m.) (c). ND, not detectable.

Extended Data Figure 2 Allo-HSCT patient timelines and C. difficile infection status transitions.

Transitions between C. difficile (_tcdB_-positive) colonization status in patients receiving allogeneic haematopoietic stem-cell transplantation, as measured by C. difficile 16S rRNA abundance during the period of hospitalization (light grey bars). Time points when C. difficile colonization was determined to be positive (red diamonds) and negative (blue diamonds), and when C. difficile infection was clinically diagnosed (black dots) and metronidazole was administered (dark grey bars), are displayed relative to the time of transplantation per patient.

Extended Data Figure 3 Identification of bacteria conserved across human and murine intestinal microbiota predicted to inhibit C. difficile.

Identification of bacterial OTUs abundant in mice (n = 68) and humans (n = 24) (a) that account for a minority of OTU membership (b) but the majority of the structure of the intestinal microbiota of both host species after antibiotic exposure (c). Subnetworks of abundant OTUs predicted inhibit (blue) or positively associate with (red) C. difficile in murine (d) and human (e) intestinal microbiota.

Extended Data Figure 4 Phylogenetic distribution of resistance-associated intestinal bacteria and isolates selected for adoptive transfer.

The maximum likelihood phylogenetic tree (Kimura model, bootstrap of 100 replicates) was constructed using the MEGA 6.06 package from representative sequences of intestinal bacteria associated with resistance to C. difficile infection (blue), including cultured representatives subsequently used in adoptive transfer experiments (bold). The tree was rooted using intestinal bacteria associated with susceptibility to infection (red) as an outgroup.

Extended Data Figure 5 Adoptive transfer and engraftment of four-bacteria consortium or C. scindens ameliorates intestinal C. difficile cytotoxin load and acute _C. difficile_-associated weight loss.

a, C. difficile toxin load in antibiotic-exposed animals receiving adoptive transfers 24 h after C. difficile infection challenge. Animals’ weights 48 h after infection challenge and (b) C. difficile c.f.u. 24 h after infection challenge (c). d, Engraftment of bacterial isolates in the intestinal microbiota of antibiotic-exposed animals 2 days after adoptive transfer of B. intestihominis, P. capillosus, B. hansenii, and/or C. scindens. e, Intestinal bacterial density (faeces) from antibiotic-exposed mice administered suspensions containing four bacteria, C. scindens, or vehicle (PBS) as measured by rtPCR of 16S rRNA genes. ****P < 0.0001, ***P < 0.001, **P < 0.01, *P < 0.05; Mann–Whitney (two-tailed) (a, b, d, e), Kruskal–Wallis with Dunn correction (c) (n = 6–10 per group). Centre values, mean; error bars, s.e.m. Results are representative of at least two independent experiments. Numbers under group columns in d denote the number of mice with detectable engraftment of the given bacterium (out of ten possible separately housed animals per group).

Extended Data Figure 6 Adoptive transfer of consortia or C. scindens restores baiCD and the abundance of the gene family responsible for secondary bile acid biosynthesis.

a, PCR-based detection of the 7α-HSDH-encoding baiCD gene in bacterial isolates, intestinal microbiomes (faeces) of animals before antibiotic exposure, and intestinal microbiomes (faeces) of animals that, after antibiotic exposure, remained susceptible to C. difficile or recovered resistance to infection spontaneously or after adoptive transfer of bacterial isolates. b, Reconstituted abundance of the gene family responsible for secondary bile acid biosynthesis, as predicted by PICRUSt, in antibiotic-exposed animals receiving adoptive transfers (n = 10 per group). ***P < 0.001; *P < 0.05; NS, not significant; Mann–Whitney (two-tailed) (b). Centre values, mean; error bars, s.e.m.

Extended Data Figure 7 Impacts of adoptive transfers of bacteria on intestinal abundance of bile acids.

Intestinal abundance of the secondary bile acids LCA (a), ursodeoxycholate (UDCA) (b), and primary bile acids (cf) in mice after antibiotic exposure and adoptive transfer of bacteria indicated. ****P < 0.0001, *P < 0.05, NS (not significant); Kruskal–Wallis test with Dunn’s correction. Centre values, mean; error bars, s.e.m.

Extended Data Figure 8 C. difficile growth inhibition by secondary bile acids and intestinal content from antibiotic-naive animals.

Addition of the secondary bile acids DCA (a) or LCA (b) to culture media inhibits C. difficile. Bile acid dependent inhibition of C. difficile enumerated by recovery of c.f.u. after inoculation of vegetative C. difficile into cell-free (c) or whole (d) intestinal content harvested from C57BL/6J mice (n = 5 or 6 per group), with or without pre-incubation with cholestyramine. **P < 0.01; Mann–Whitney (two-tailed) (c, d).

Extended Data Table 1 Characteristics of patients and transplant course

Full size table

Extended Data Table 2 Retention times for bile acids quantified by high-performance liquid chromatography–mass spectrometry

Full size table

PowerPoint slides

Rights and permissions

About this article

Cite this article

Buffie, C., Bucci, V., Stein, R. et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile.Nature 517, 205–208 (2015). https://doi.org/10.1038/nature13828

Download citation