Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Altered microbial bile acid metabolism exacerbates T cell-driven inflammation during graft-versus-host disease

Abstract

Microbial transformation of bile acids affects intestinal immune homoeostasis but its impact on inflammatory pathologies remains largely unknown. Using a mouse model of graft-versus-host disease (GVHD), we found that T cell-driven inflammation decreased the abundance of microbiome-encoded bile salt hydrolase (BSH) genes and reduced the levels of unconjugated and microbe-derived bile acids. Several microbe-derived bile acids attenuated farnesoid X receptor (FXR) activation, suggesting that loss of these metabolites during inflammation may increase FXR activity and exacerbate the course of disease. Indeed, mortality increased with pharmacological activation of FXR and decreased with its genetic ablation in donor T cells during mouse GVHD. Furthermore, patients with GVHD after allogeneic hematopoietic cell transplantation showed similar loss of BSH and the associated reduction in unconjugated and microbe-derived bile acids. In addition, the FXR antagonist ursodeoxycholic acid reduced the proliferation of human T cells and was associated with a lower risk of GVHD-related mortality in patients. We propose that dysbiosis and loss of microbe-derived bile acids during inflammation may be an important mechanism to amplify T cell-mediated diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: T cell-driven inflammation alters the intestinal BA pool.
Fig. 2: Alloreactive T cells alter host and microbial BA metabolism.
Fig. 3: FXR activity contributes to GVHD.
Fig. 4: Comparison of BA pools in patients with GVHD vs controls.
Fig. 5: BA biotransformation potential in transplant patients with GVHD vs controls.
Fig. 6: UDCA limits human effector T cell responses and is associated with improved GVHD-related mortality.

Similar content being viewed by others

Data availability

Metabolomics data including standards (Figs. 1, 4 and 5, and Extended Data Figs. 47 and Tables 4 and 5) are available at GNPS (https://gnps.ucsd.edu/) under MassIVE project ID #MSV000092300. The bulk RNA-seq data from murine experiments (Fig. 2 and Extended Data Fig. 2) are available at NCBI GEO under GEO accession GSE218343 and also in Supplementary tables. The 16S and shotgun sequencing data (Figs. 2 and 5, and Extended Data Fig. 7) are available at NCBI under accession numbers listed in the Supplementary tables. The processed scRNA-seq files are available under GEO accession GSE253360. For access to raw data, kindly request permission by contacting the contributing author at mvandenbrink@coh.org. Please anticipate a response within 2 weeks. Once legal agreements are approved, raw genomic data can be shared within an additional month. Source data for Figs. 13 and 6ac, and for Extended Data Figs. 13 and 8 are available in the Supplementary tables. The datasets required to run the code were also made publicly available in GitHub and are also included in the Supplementary information. Our institutional data-sharing policies prevent us from publicly posting the patient-level information used to calculate clinical outcomes (Fig. 6t,u). However, interested parties may request access by contacting the contributing author at mvandenbrink@coh.org. Please anticipate a response within one month. Data sharing of patient-level information is contingent upon the establishment of a formal data transfer agreement between Memorial Sloan Kettering and the respective parties involved. Source data are provided with this paper.

Code availability

The code and the corresponding figures for Figs. 2d,e, 4, 5 and 6h–v, and for Extended Data Figs. 47 and 10 can be accessed in GitHub (https://github.com/orianamiltiadous/BAs_and_GVHD).

References

  1. Hamilton, J. P. et al. Human cecal bile acids: concentration and spectrum. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G256–G263 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Chen, M. L., Takeda, K. & Sundrud, M. S. Emerging roles of bile acids in mucosal immunity and inflammation. Mucosal Immunol. 12, 851–861 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Belkaid, Y. & Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 157, 121–141 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Russell, D. W. The enzymes, regulation, and genetics of bile acid synthesis. Annu. Rev. Biochem. 72, 137–174 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. 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  PubMed  Google Scholar 

  6. Begley, M., Hill, C. & Gahan, C. G. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 72, 1729–1738 (2006).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  7. Wells, J. E. & Hylemon, P. B. Identification and characterization of a bile acid 7alpha-dehydroxylation operon in Clostridium sp. strain TO-931, a highly active 7alpha-dehydroxylating strain isolated from human feces. Appl. Environ. Microbiol. 66, 1107–1113 (2000).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  8. Devlin, A. S. & Fischbach, M. A. A biosynthetic pathway for a prominent class of microbiota-derived bile acids. Nat. Chem. Biol. 11, 685–690 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Quinn, R. A. et al. Global chemical effects of the microbiome include new bile-acid conjugations. Nature 579, 123–129 (2020).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  10. Foley, M. H. et al. Bile salt hydrolases shape the bile acid landscape and restrict Clostridioides difficile growth in the murine gut. Nat. Microbiol. 8, 611–628 (2023).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Patterson, A. et al. Bile acids are substrates for amine n-acyl transferase activity by bile salt hydrolase. Preprint at Res. Square https://doi.org/10.21203/rs.3.rs-2050120/v1 (2022).

  12. Shin, D. J. & Wang, L. Bile acid-activated receptors: a review on FXR and other nuclear receptors. Handb. Exp. Pharmacol. 256, 51–72 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Hang, S. et al. Bile acid metabolites control T(H)17 and T(reg) cell differentiation. Nature 576, 143–148 (2019).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  14. Paik, D. et al. Human gut bacteria produce Τ(Η)17-modulating bile acid metabolites. Nature 603, 907–912 (2022).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  15. Campbell, C. et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 581, 475–479 (2020).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  16. Gratwohl, A. et al. Hematopoietic stem cell transplantation: a global perspective. JAMA 303, 1617–1624 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Gooley, T. A. et al. Reduced mortality after allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 363, 2091–2101 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Aljurf, M. et al. Worldwide Network for Blood & Marrow Transplantation (WBMT) special article, challenges facing emerging alternate donor registries. Bone Marrow Transpl. 54, 1179–1188 (2019).

    Article  Google Scholar 

  19. Peled, J. U. et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 382, 822–834 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Taur, Y. et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 124, 1174–1182 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Jenq, R. R. et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J. Exp. Med. 209, 903–911 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Holler, E. et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol. Blood Marrow Transpl. 20, 640–645 (2014).

    Article  Google Scholar 

  23. Burgos da Silva, M. et al. Preservation of fecal microbiome is associated with reduced severity of graft-versus-host disease. Blood 140, 2385–2397 (2022).

  24. 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  PubMed Central  PubMed  Google Scholar 

  25. Ara, T. & Hashimoto, D. Novel insights into the mechanism of GVHD-induced tissue damage. Front. Immunol. 12, 713631 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Zeiser, R. & Blazar, B. R. Acute graft-versus-host disease – biologic process, prevention, and therapy. N. Engl. J. Med. 377, 2167–2179 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Malard, F., Holler, E., Sandmaier, B. M., Huang, H. & Mohty, M. Acute graft-versus-host disease. Nat. Rev. Dis. Prim. 9, 27 (2023).

    Article  PubMed  Google Scholar 

  28. Reddy, P., Negrin, R. & Hill, G. R. Mouse models of bone marrow transplantation. Biol. Blood Marrow Transpl. 14, 129–135 (2008).

    Article  Google Scholar 

  29. Palmer, R. H. The formation of bile acid sulfates: a new pathway of bile acid metabolism in humans. Proc. Natl Acad. Sci. USA 58, 1047–1050 (1967).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  30. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  31. Mootha, V. K. et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Gadaleta, R. M. et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 60, 463–472 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Campbell, C. et al. FXR mediates T cell-intrinsic responses to reduced feeding during infection. Proc. Natl Acad. Sci. USA 117, 33446–33454 (2020).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  34. Sayin, S. I. et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 17, 225–235 (2013).

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  35. Shono, Y. et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci. Transl. Med. 8, 339ra371 (2016).

    Article  ADS  Google Scholar 

  36. Ruutu, T. et al. Ursodeoxycholic acid for the prevention of hepatic complications in allogeneic stem cell transplantation. Blood 100, 1977–1983 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Ruutu, T. et al. Improved survival with ursodeoxycholic acid prophylaxis in allogeneic stem cell transplantation: long-term follow-up of a randomized study. Biol. Blood Marrow Transpl. 20, 135–138 (2014).

    Article  CAS  Google Scholar 

  38. Guzior, D. V. & Quinn, R. A. Review: microbial transformations of human bile acids. Microbiome 9, 140 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Claudel, T., Staels, B. & Kuipers, F. The Farnesoid X receptor: a molecular link between bile acid and lipid and glucose metabolism. Arterioscler Thromb. Vasc. Biol. 25, 2020–2030 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Cox, D. R. Regression models and life-tables. J. R. Stat. Soc. B 34, 187–202 (1972).

    MathSciNet  Google Scholar 

  41. Fine, J. P. & Gray, R. J. A proportional hazards model for the subdistribution of a competing risk. Theory Method 94, 496–509 (1997).

  42. Haring, E. et al. Bile acids regulate intestinal antigen presentation and reduce graft-versus-host disease without impairing the graft-versus-leukemia effect. Haematologica 106, 2131–2146 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Gevers, D. et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 15, 382–392 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Peled, J. U. et al. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J. Clin. Oncol. 35, 1650–1659 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  45. McCarville, J. L., Chen, G. Y., Cuevas, V. D., Troha, K. & Ayres, J. S. Microbiota metabolites in health and disease. Annu. Rev. Immunol. 38, 147–170 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Yao, L. et al. A selective gut bacterial bile salt hydrolase alters host metabolism. Elife 7, e37182 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  47. Zhang, Y. et al. Ursodeoxycholic acid accelerates bile acid enterohepatic circulation. Br. J. Pharmacol. 176, 2848–2863 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  48. Winston, J. A., Rivera, A., Cai, J., Patterson, A. D. & Theriot, C. M. Secondary bile acid ursodeoxycholic acid alters weight, the gut microbiota, and the bile acid pool in conventional mice. PLoS ONE 16, e0246161 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Song, X. et al. Microbial bile acid metabolites modulate gut RORγ(+) regulatory T cell homeostasis. Nature 577, 410–415 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Li, W. et al. A bacterial bile acid metabolite modulates T(reg) activity through the nuclear hormone receptor NR4A1. Cell Host Microbe 29, 1366–1377.e9 (2021).

  51. Lee, J. W. J. et al. Multi-omics reveal microbial determinants impacting responses to biologic therapies in inflammatory bowel disease. Cell Host Microbe 29, 1294–1304.e4 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  53. Smith, M. et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat. Med. 28, 713–723 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  54. Stein-Thoeringer, C. K. et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science 366, 1143–1149 (2019).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  55. Mathewson, N. D. et al. Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat. Immunol. 17, 505–513 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  57. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

    MathSciNet  Google Scholar 

  58. Andrews, S. FastQC v.0.11.9 (Babraham Bioinformatics, 2015).

  59. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  60. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

  61. Peled, J. U., Gomes, A. L. C. & van den Brink, M. R. M. Microbiota and allogeneic hematopoietic-cell transplantation. Reply. N. Engl. J. Med. 382, 2378–2379 (2020).

    Article  PubMed  Google Scholar 

  62. Miltiadous, O. et al. Early intestinal microbial features are associated with CD4 T-cell recovery after allogeneic hematopoietic transplant. Blood 139, 2758–2769 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Callahan, B. J., McMurdie, P. J. & Holmes, S. P. Exact sequence variants should replace operational taxonomic units in marker-gene data analysis. ISME J. 11, 2639–2643 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  64. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  65. Dubin, K. A. et al. Diversification and evolution of vancomycin-resistant Enterococcus faecium during intestinal domination. Infect. Immun. 87, e00102–e00119 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  66. Beghini, F. et al. Integrating taxonomic, functional, and strain-level profiling of diverse microbial communities with bioBakery 3. Elife 10, e65088 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  67. Buchfink, B., Xie, C. & Huson, D. H. Fast and sensitive protein alignment using DIAMOND. Nat. Methods 12, 59–60 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Buchfink, B., Reuter, K. & Drost, H. G. Sensitive protein alignments at tree-of-life scale using DIAMOND. Nat. Methods 18, 366–368 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  69. Menzel, P., Ng, K. L. & Krogh, A. Fast and sensitive taxonomic classification for metagenomics with Kaiju. Nat. Commun. 7, 11257 (2016).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  70. Stoeckius, M. et al. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  71. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  72. Kousa, A. I. & Lemarquis, A. The shunPykeR’s guide to single cell analysis (v.1.0.0). GitHub https://github.com/kousaa/shunPykeR?tab=readme-ov-file (2023).

  73. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  ADS  CAS  PubMed Central  PubMed  Google Scholar 

  74. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  76. Ntranos, V., Yi, L., Melsted, P. & Pachter, L. A discriminative learning approach to differential expression analysis for single-cell RNA-seq. Nat. Methods 16, 163–166 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge R. Chaligne, the single-cell analytics innovation laboratory (SAIL), the Integrated Genomics Operation Core (IGO) and the molecular microbiology facility (MMF), which performed RNA sequencing (SAIL, IGO), as well as the 16S and metagenomic shotgun sequencing (MMF, IGO) for mouse and human studies. This research is funded by the National Cancer Institute (NCI) Cancer Center Support Grant (CCSG, P30 CA08748), Cycle for Survival, and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology NCI award numbers R01-CA228358, R01-CA228308, P30 CA008748 MSK Cancer Center Support Grant/Core Grant and P01-CA023766; National Heart, Lung and Blood Institute (NHLBI) award number R01-HL123340 and R01-HL147584; and the Tri Institutional Stem Cell Initiative. Additional funding was received from The Lymphoma Foundation, The Susan and Peter Solomon Family Fund, The Solomon Microbiome Nutrition and Cancer Program, Cycle for Survival, Parker Institute for Cancer Immunotherapy, Paula and Rodger Riney Multiple Myeloma Research Initiative, Starr Cancer Consortium, and Seres Therapeutics. S.L. was supported by the Deutsche Forschungsgemeinschaft (DFG, LI 3565/1-1) and DKMS. O.M. was supported by the American Society of Clinical Oncology Young Investigator Award, a Hyundai Hope on Wheels Young Investigator Award, a Cycle for Survival Equinox Innovation Award, a Collaborative Pediatric Cancer Research Program Award, a Michael Goldberg Fellowship and a Tow Center for Developmental Oncology Career Development Award. K.A.M. was supported by the DKMS and the ASH Scholar Award. J.U.P. reports funding from NHLBI NIH Award K08HL143189.

Author information

Authors and Affiliations

Authors

Contributions

S.L., O.M., C.C. and M.R.M.v.d.B designed the study and wrote the manuscript. S.L. and C.C. performed harvest experiments and flow cytometric analyses. S.L. performed BM + T experiments and FXR reporter assays. O.M. selected the patient cohort and analysed human bile acid profiling and metagenomic data. R.J.F.R. and J.R.C. quantified and analysed the LC–MS/MS bile acid profiling data and advised on study design. A.I.K. analysed the mouse RNA and human scRNA-seq data. A.D. analysed the mouse metagenomic sequencing data. N.R.W. assisted with code resources and oversight. K.S. assisted with organizing the sequencing files and creating biorepositories. J.P. performed human and mice in vitro T cell assays and flow cytometric analysis. T.F. performed biostatistical analysis. E.L. and P.R. performed ΔFXR recipient BM + T. J.F. assisted with the analysis of metagenomic data. G.K.A., R.G., K.V. and B.G. assisted with BM + T experiments. S.M. carried out histopathological analyses of tissues. J.S. coordinated the faecal microbiome collection. M.V.R., C.L.N., Y.T., K.A.M., H.A., M.B.d.S., J.U.P. and M.S. contributed to analysis strategies. S.G. and M.-A.P. contributed to clinical data collection. C.C. and M.R.M.v.d.B. supervised the study and contributed equally. S.L. and O.M. contributed equally. All authors reviewed and approved the manuscript.

Corresponding authors

Correspondence to Clarissa Campbell or Marcel R. M. van den Brink.

Ethics declarations

Competing interests

M.-A.P. reports honoraria from Adicet, Allovir, Caribou Biosciences, Celgene, Bristol-Myers Squibb, Equilium, Exevir, Incyte, Karyopharm, Kite/Gilead, Merck, Miltenyi Biotec, MorphoSys, Nektar Therapeutics, Novartis, Omeros, OrcaBio, Syncopation, VectivBio AG and Vor Biopharma; he serves on DSMBs for Cidara Therapeutics, Medigene and Sellas Life Sciences, and the scientific advisory board of NexImmune; he has ownership interests in NexImmune and Omeros; and he has received institutional research support for clinical trials from Incyte, Kite/Gilead, Miltenyi Biotec, Nektar Therapeutics and Novartis. K.A.M. holds equity and is on the advisory board of Postbiotics Plus, and has consulted for Incyte. J.U.P. reports research funding, intellectual property fees and travel reimbursement from Seres Therapeutics, and consulting fees from Da Volterra, CSL Behring and MaaT Pharma; he serves on the Advisory board of and holds equity in Postbiotics Plus Research; and he has filed intellectual property applications related to the microbiome (reference numbers 62/843,849, 62/977,908 and 15/756,845). M.R.M.v.d.B. has received research support and stock options from Seres Therapeutics, and stock options from Notch Therapeutics and Pluto Therapeutics; he has received royalties from Wolters Kluwer; he has consulted, received honorarium from or participated in advisory boards for Seres Therapeutics, Vor Biopharma, Rheos Medicines, Frazier Healthcare Partners, Nektar Therapeutics, Notch Therapeutics, Ceramedix, Lygenesis, Pluto Therapeutics, GlaskoSmithKline, Da Volterra, Thymofox, Garuda, Novartis (Spouse), Synthekine (Spouse), Beigene (Spouse) and Kite (Spouse); he has IP Licensing with Seres Therapeutics and Juno Therapeutics; and he holds a fiduciary role on the Foundation Board of DKMS (a nonprofit organization). Memorial Sloan Kettering has institutional financial interests relative to Seres Therapeutics. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Pieter Dorrestein, Kenya Honda and Olle Ringden for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Impact of alloreactive T cells on BA ratios and clinical markers.

Lethally irradiated 6-8 week old female BALBc mice were transplanted with 10 ×106 B6 BM cells alone (BM) or together with 1 ×106 T cells (BM + T). BAs were quantified on day 7 post-transplant by LC-MS in the cecal contents and plasma: (a) ratio of microbe- to host-derived BAs, (b) ratio of unconjugated to glycine- and taurine-conjugated BA and (c) estimated cecal levels of the T cell modulatory BAs 3-oxoLCA and isoLCA (below the linear range of quantification. Weight loss (d), clinical GVHD scores (e) and cumulative food intake per mouse (f), plasma levels of AST (g), ALT (h), albumin (i), cholesterol (j), and triglycerides (k) at day 7 post-transplant of these mice. (a, b, d-k) Data combined from two independent experiments (n = 10). (c) Data is representative of two independent experiments (n = 5). (a-k) Data shown as mean ± S.D and statistical significance determined by two-tailed Mann-Whitney test.

Extended Data Fig. 2 FXR Signaling in T cells modulates GVHD.

(a) Principle of the FXR luciferase reporter assay used in Fig. 3a and b and Extended Data Fig. 2b. (b) Stably transfected HepG2 cells expressing luciferase under the control of an FXR-responsive element were treated with the indicated doses of CDCA. Luciferase units (luminescence) were normalized to cell viability assessed by Hoechst 33342 staining (fluorescence). Data representative of two independent experiments and presented as technical triplicates with means connected. Weight loss (c) and clinical GVHD score (d) of survival experiment shown in Fig. 3d. Data combined from three independent experiments (BM group n = 20, BM + T groups n = 30 per group) and means ± S.D connected. (e) Survival of cohoused WT or nr1h4-/-l (Δ FXR) B6 mice receiving BALBc BM + T. Data combined from three independent experiments (n = 13 per group). Statistical significance was determined using log-rank test. (f-h) BALBc recipient mice transplanted with 10 ×106 B6 BM cells alone or together with 1 ×106 T cells from either Nr1h4fl/fl (BM + TWT) or Cd4Cre Nr1h4fl/fl (BM + TΔFXR) mice on a C57Bl/6 N background. (f) Organ-specific and compound histopathological scores at day 28 post-transplant of transplanted mice with representative histology images shown in (g). Data from one experiment (n = 10 per group) and statistical significance was determined by two-tailed Mann-Whitney test. (h) Production of IFNγ by CD4+ and CD8+ T cells in the smallI and large intestine lamina propria 14 days after transplant. Data combined from two (n = 10 per group) and presented as mean ± S.D. Statistical significance was determined by by two-tailed Mann-Whitney test.

Extended Data Fig. 3 Consort diagram.

Criteria for the cohort selection used for data shown in Figs. 4, 5 and Extended Data Fig. 5-8. PBSC: peripheral blood stem cell graft.

Extended Data Fig. 4 Effects of UDCA exposure on the intestinal BA pool.

Data from n = 280 samples from either peri-engraftment or peri-GVHD onset timepoints. Fecal concentrations of UDCA (a) and microbe-derived BAs (b). UDCA exposure status is shown in the x-axis (w = weeks; m = months since last exposure). Statistical significance determined by the 2-sided Wilcoxon Rank-sum test. The boxplot center line corresponds to the median, box limits correspond to the 25th and 75th percentile, and whiskers correspond to 1.5x interquartile range. Correlation of fecal UDCA concentrations with the levels of conjugated UDCA (conj-UDCA, c), total BAs (d), microbe-derived BAs (e), host-derived BAs (f), nonUDCA total BAs (g) nonUDCA microbe-derived BAs (h), and microbe- to host- derived (M/H) ratio excluding UDCA (nonUDCA, i). The solid line represents a linear regression model fitted to the data. The shaded region surrounding the line indicates a 95% confidence interval for the regression line. Total BAs nonUDCA (g) are measured in pmol/mg. (j,k) Correlation matrix of the BA species covarying with UDCA. (k) Showing BA species with a Pearson correlation coefficient (R > 0.4).

Extended Data Fig. 5 Fecal BA profiles at the peri-GVHD onset time point.

Showing the levels of total microbe-derived BAs (a), microbe- to host-derived (M/H) BA ratio (b), M*/H (that is, nonUDCA M/H) BA ratio (c), and the ratio of unconjugated to amidated BAs (d). Data representative of 57 control and 58 GVHD patients. Statistical significance determined with the 2-sided Wilcoxon Rank-sum test. The boxplot center line corresponds to the median, box limits correspond to the 25th and 75th percentile, and whiskers correspond to 1.5x interquartile range. (e-f) Differential abundance of BAs between GVHD and controls in peri-GVHD onset samples after multivariate adjustment. (e) Grid plot showing significance status, q-values, and log fold changes of BAs relative to indicated clinical variables. (f) Volcano plot showing log-transformed adjusted p-values vs log fold changes of BAs between GVHD and control patients. Statistical comparison was made using the two-sided empirical Bayes moderated t-test and p-values were adjusted using the Benjamini-Hochberg method.

Extended Data Fig. 6 Fecal BA profiles at the peri-engraftment time point.

Total BAs (a), host-derived (b), microbe-derived (c), microbe*-derived (d), microbe-derived to host-derived (M/H) BA ratio (e), M*/H (nonUDCA M/H) BA ratio in patients that develop GVHD vs controls (f). (g) Pie chart showing the averaged relative contributions of host-derived and microbe*-derived to the calculated total BA pool. Glycine- and taurine-conjugated (h), unconjugated (i), and sulfated (g) BAs. Pie chart showing the averaged percentages of glycine- and taurine-conjugated, unconjugated and sulfated BAs in patients with GVHD vs controls in peri-GVHD onset samples (k). Microbe*-derived BAs: Microbe-derived BAs excluding UDCA. Data representative of 90 control and 86 GVHD patients. The boxplot center line corresponds to the median, box limits correspond to the 25th and 75th percentile, and whiskers correspond to 1.5x interquartile range. Statistical significance determined with the 2-sided Wilcoxon Rank-sum test. (l,m) Differential abundance of BAs between GVHD and control patients in peri-engraftment samples after multivariate adjustment (l) Grid plot showing significance status, q-values, and log fold changes of BAs relative to indicated clinical variables. (m) Volcano plot showing log-transformed adjusted p-values vs log fold changes of BAs between GVHD and control patients. Statistical comparison was made using the two-sided empirical Bayes moderated t-test and p-values were adjusted using the Benjamini-Hochberg method.

Extended Data Fig. 7 Microbiome features in peri-GVHD onset samples.

Relative abundance of (a) Eggerthella lenta and (b) Ruminococcus gnavus. Data representative of 49 control and 46 GVHD patients (c) Correlation of the sum of bai operon gene and α-diveristy as measured by the Simpson reciprocal index. The solid line represents a linear regression model fitted to the data. The shaded region surrounding the line indicates a 95% confidence interval for the regression line. Data representative of 82 patients with peri-GVHD onset samples. (d) α-diversity, (e) sum of bai operon genes identified by shotgun metagenomic analysis (measured in counts per million), and (f) levels of microbe-derived BAs* (pmol/mg) in patients with or without intestinal pathogen domination. Data representative of 41 patients with and 74 patients without pathogen domination. Microbe-derived BAs*= microbe-derived BAs excluding UDCA. Statistical significance determined with the univariate 2-sided Wilcoxon Rank-sum test. The boxplot center line corresponds to the median, box limits correspond to the 25th and 75th percentile, and whiskers correspond to 1.5x interquartile range. R correspond to Pearson’s correlation coefficient.

Extended Data Fig. 8 In vitro human T cell proliferation in response to FXR activation or inhibition with drugs or BAs.

(a) Experimental design. Purified human T cells were activated with anti-CD3 and anti-CD28 antibodies in the presence of recombinant IL-2 for 2 days and further cultured either in the presence of anti-CD3/anti-CD28 antibodies and IL-2 (continuous activation control) or in their absence (vehicle control) with or without the indicated compounds for 96 hours. Showing T cell confluence in response to CDCA and UDCA (b) or GW4064 and DY268 (c) at the indicated concentrations. Cell viability (d, e, f) and representative histograms (g, h) showing CD25 levels determined by flow cytometric analysis. (i-l) CD25 expression in CD4+ and CD8+ T cells after 96 hours of treatment with CDCA and UDCA (i-j), or GW4064 and DY268 (k-l). Showing the geometric mean fluorescence intensity (MFI) of CD25 in CD25+ T cells. Values were normalized to the MFI of the vehicle-treated group. (m, n) Frequency of CD25 positive cells on day 4 post-activation. (o) CD25 expression from T cells of FXRWT or FXRWT mice treated with anti-CD3 and anti-CD28 antibodies in the presence of IL-2 for 2 days before incubation with CDCA (100 nM), or UDCA (100 nM), anti-CD3, anti-CD28 and IL-2 (continuous activation), or vehicle for 2 more days. CD25 expression was measured as geometric mean fluorescence intensity (MFI) of CD25 in CD25+ T cells normalized to the MFI of the vehicle-treated group. Statistical analysis was performed by two-way (b,c) or one-way ANOVA followed by multiple t-test with Bonferoni correction (d-f, i-o). Each data point in (i-n) shows the average of technical duplicates for a single donor. Bars denote the standard error of the mean. Data representative of 4 independent experiments with a total of 4 PBMC donors. Each data point in (g) shows the average of technical triplicates from two mice. Bars denote the standard error of the mean. Data representative of 3 independent experiments with a total of 6 mice.

Extended Data Fig. 9 Quality control of single cell RNA-sequencing profiling of in vitro activated T cells treated with FXR ligands, DMSO or activating signals for 24 h.

Visualization of 60,767 cells using a uniform manifold approximation and projection (UMAP) of (a) cells from the two donors and (b) per hashtag before eliminating any cells. (c) Total counts (log10 scale) (d) total genes, (e) ribosomal fraction, (f) mitochondrial fraction per cell, (g) predicted doublet and (h) doublet score. (i-k) Cells expressing markers for B and Natural Killer cells were defined as contaminants. (l-m) UMAP and stacked plot showing the fraction of retained (33,634) and removed cells (27,133).

Extended Data Fig. 10 Single cell RNA-sequencing profiling of in vitro activated T cells treated with CDCA (100 nM), UDCA (100 nM), GW4064 (1uM) and DY268 (1uM) for 24 h.

(a) Gene markers used to identify cell populations. Visualization of annotated cells using a uniform manifold approximation and projection (UMAP) of (b) subtypes after batch correction and (c) per treatment arm. (d) Gene Set Enrichment Analysis of pathways differentially regulated in the different conditions (CDCA 100 nM, UDCA 100 nM, GW4064 1uM, DY268 1uM) relative to the vehicle control in CD4+, CD8+ and regulatory T cell populations. Displaying significant pathways.

Supplementary information

Supplementary Information

Dot plot for metabolomics in mice, gating strategy and Supplementary Tables 1–9.

Reporting Summary

Peer Review File

Supplementary Table

Bulk RNA-sequencing of liver, small and large intestine; source data for extended data figures; ASV sequences; antibody dilutions.

Source data

Source Data Fig. 1

BA levels in mice with GVHD vs controls

Source Data Fig. 2

Abundance of BA-related genes and results of bulk RNA-sequencing of liver tissue and epithelial fractions of the small and large intestines in mice with GVHD vs controls.

Source Data Fig. 3

Transcriptional activity of FXR in response to treatment with individual bile acids; survival outcomes associated with enhanced FXR activity and FXR knockout (KO) conditions.

Source Data Fig. 4

Information on the cohort (GVHD vs controls), the concentrations/AUC of BAs, BA family.

Source Data Fig. 5

Abundance of BA-related genes, α-diversity, ASV counts used for the composition plot.

Source Data Fig. 6

T cell confluence in response to different treatment arms; CD25 MFI in response to different treatment arms; source data for fold-change plots in CD4 T cells.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lindner, S., Miltiadous, O., Ramos, R.J.F. et al. Altered microbial bile acid metabolism exacerbates T cell-driven inflammation during graft-versus-host disease. Nat Microbiol 9, 614–630 (2024). https://doi.org/10.1038/s41564-024-01617-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-024-01617-w

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research