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:

Self-tunable engineered yeast probiotics for the treatment of inflammatory bowel disease

Abstract

Inflammatory bowel disease (IBD) is a complex chronic inflammatory disorder of the gastrointestinal tract. Extracellular adenosine triphosphate (eATP) produced by the commensal microbiota and host cells activates purinergic signaling, promoting intestinal inflammation and pathology. Based on the role of eATP in intestinal inflammation, we developed yeast-based engineered probiotics that express a human P2Y2 purinergic receptor with up to a 1,000-fold increase in eATP sensitivity. We linked the activation of this engineered P2Y2 receptor to the secretion of the ATP-degrading enzyme apyrase, thus creating engineered yeast probiotics capable of sensing a pro-inflammatory molecule and generating a proportional self-regulated response aimed at its neutralization. These self-tunable yeast probiotics suppressed intestinal inflammation in mouse models of IBD, reducing intestinal fibrosis and dysbiosis with an efficacy similar to or higher than that of standard-of-care therapies usually associated with notable adverse events. By combining directed evolution and synthetic gene circuits, we developed a unique self-modulatory platform for the treatment of IBD and potentially other inflammation-driven pathologies.

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

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

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

Fig. 1: Directed evolution of the human P2Y2 receptor.
Fig. 2: Characterization of human P2Y2 receptor mutants.
Fig. 3: eATP-responsive secretion of ATPase by engineered yeast.
Fig. 4: eATP-responsive engineered yeasts ameliorate chemically induced colitis.
Fig. 5: eATP-responsive engineered yeasts ameliorate anti-CD3 antibody-induced enteritis.
Fig. 6: eATP-responsive engineered yeast probiotics limit fibrosis and dysbiosis.

Similar content being viewed by others

Data availability

RNA-seq data were deposited in the GEO database under the accession number GSE152869. Sequencing data from microbiota 16S rRNA were submitted to the NCBI short-read archive under BioProject number PRJNA641709. Source data are provided with this paper.

References

  1. Huang, H. et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 547, 173–178 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Neurath, M. F. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat. Immunol. 20, 970–979 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Blander, J. M., Longman, R. S., Iliev, I. D., Sonnenberg, G. F. & Artis, D. Regulation of inflammation by microbiota interactions with the host. Nat. Immunol. 18, 851–860 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jostins, L. et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Plichta, D. R., Graham, D. B., Subramanian, S. & Xavier, R. J. Therapeutic opportunities in inflammatory bowel disease: mechanistic dissection of host–microbiome relationships. Cell 178, 1041–1056 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Suez, J., Zmora, N., Segal, E. & Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 25, 716–729 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Aggarwal, N., Breedon, A. M. E., Davis, C. M., Hwang, I. Y. & Chang, M. W. Engineering probiotics for therapeutic applications: recent examples and translational outlook. Curr. Opin. Biotechnol. 65, 171–179 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Hwang, I. Y. et al. Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models. Nat. Commun. 8, 15028 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. McKay, R. et al. A platform of genetically engineered bacteria as vehicles for localized delivery of therapeutics: toward applications for Crohn’s disease. Bioeng. Transl. Med. 3, 209–221 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Idzko, M., Ferrari, D. & Eltzschig, H. K. Nucleotide signalling during inflammation. Nature 509, 310–317 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Atarashi, K. et al. ATP drives lamina propria TH17 cell differentiation. Nature 455, 808–812 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Gulbransen, B. D. et al. Activation of neuronal P2X7 receptor–pannexin-1 mediates death of enteric neurons during colitis. Nat. Med. 18, 600–604 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Takenaka, M. C., Robson, S. & Quintana, F. J. Regulation of the T cell response by CD39. Trends Immunol. 37, 427–439 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mascanfroni, I. D. et al. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39. Nat. Immunol. 14, 1054–1063 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Takenaka, M. C. et al. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat. Neurosci. 22, 729–740 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cekic, C. & Linden, J. Purinergic regulation of the immune system. Nat. Rev. Immunol. 16, 177–192 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Vuerich, M., Robson, S. C. & Longhi, M. S. Ectonucleotidases in intestinal and hepatic inflammation. Front. Immunol. 10, 507 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sen, S. & Mansell, T. J. Yeasts as probiotics: mechanisms, outcomes, and future potential. Fungal Genet. Biol. 137, 103333 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Brown, A. J. et al. Functional coupling of mammalian receptors to the yeast mating pathway using novel yeast/mammalian G protein α-subunit chimeras. Yeast 16, 11–22 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Shaw, W. M. et al. Engineering a model cell for rational tuning of GPCR signaling. Cell 177, 782–796 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lazarowski, E. R. & Harden, T. K. Quantitation of extracellular UTP using a sensitive enzymatic assay. Br. J. Pharmacol. 127, 1272–1278 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wan, P. et al. Extracellular ATP mediates inflammatory responses in colitis via P2 × 7 receptor signaling. Sci. Rep. 6, 19108 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Di Virgilio, F., Pinton, P. & Falzoni, S. Assessing extracellular ATP as danger signal in vivo: the pmeLuc system. Methods Mol. Biol. 1417, 115–129 (2016).

    Article  PubMed  CAS  Google Scholar 

  25. Hillmann, P. et al. Key determinants of nucleotide-activated G protein-coupled P2Y2 receptor function revealed by chemical and pharmacological experiments, mutagenesis and homology modeling. J. Med. Chem. 52, 2762–2775 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Schutz, M. et al. Directed evolution of G protein-coupled receptors in yeast for higher functional production in eukaryotic expression hosts. Sci. Rep. 6, 21508 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Montaner, S., Kufareva, I., Abagyan, R. & Gutkind, J. S. Molecular mechanisms deployed by virally encoded G protein-coupled receptors in human diseases. Annu. Rev. Pharmacol. Toxicol. 53, 331–354 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Venkatakrishnan, A. J. et al. Diverse activation pathways in class A GPCRs converge near the G-protein-coupling region. Nature 536, 484–487 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Komoszynski, M. A. Comparative studies on animal and plant apyrases (ATP diphosphohydrolase EC 3.6.1.5) with application of immunological techniques and various ATPase inhibitors. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 113, 581–591 (1996).

    Article  CAS  PubMed  Google Scholar 

  30. Nourizad, N., Ehn, M., Gharizadeh, B., Hober, S. & Nyren, P. Methylotrophic yeast Pichia pastoris as a host for production of ATP-diphosphohydrolase (apyrase) from potato tubers (Solanum tuberosum). Protein Expr. Purif. 27, 229–237 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Derkx, B. et al. Tumour-necrosis-factor antibody treatment in Crohn’s disease. Lancet 342, 173–174 (1993).

    Article  CAS  PubMed  Google Scholar 

  32. Dotan, I. et al. The role of integrins in the pathogenesis of inflammatory bowel disease: approved and investigational anti-integrin therapies. Med. Res. Rev. 40, 245–262 (2020).

    Article  CAS  PubMed  Google Scholar 

  33. Shen, C. et al. Remission-inducing effect of anti-TNF monoclonal antibody in TNBS colitis: mechanisms beyond neutralization? Inflamm. Bowel Dis. 13, 308–316 (2007).

    Article  PubMed  Google Scholar 

  34. Goettel, J. A. et al. AHR activation is protective against colitis driven by T cells in humanized mice. Cell Rep. 17, 1318–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nowarski, R., Jackson, R. & Flavell, R. A. The stromal intervention: regulation of immunity and inflammation at the epithelial–mesenchymal barrier. Cell 168, 362–375 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Sonnenberg, G. F., Fouser, L. A. & Artis, D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat. Immunol. 12, 383–390 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Trivedi, P. P. & Jena, G. B. Dextran sulfate sodium-induced ulcerative colitis leads to increased hematopoiesis and induces both local as well as systemic genotoxicity in mice. Mutat. Res. 744, 172–183 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Musch, M. W. et al. T cell activation causes diarrhea by increasing intestinal permeability and inhibiting epithelial Na+/K+-ATPase. J. Clin. Invest. 110, 1739–1747 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ueno, N. et al. TU-100 (Daikenchuto) and ginger ameliorate anti-CD3 antibody induced T cell-mediated murine enteritis: microbe-independent effects involving Akt and NF-κB suppression. PLoS ONE 9, e97456 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Goretsky, T. et al. p53 mediates TNF-induced epithelial cell apoptosis in IBD. Am. J. Pathol. 181, 1306–1315 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. He, Q. et al. Dysbiosis of the fecal microbiota in the TNBS-induced Crohn’s disease mouse model. Appl. Microbiol. Biotechnol. 100, 4485–4494 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Frank, D. N. et al. Molecular–phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl Acad. Sci. USA 104, 13780–13785 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Machiels, K. et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 63, 1275–1283 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Chen, Y. et al. Genetic circuit design automation for yeast. Nat. Microbiol. 5, 1349–1360 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Braat, H. et al. A phase I trial with transgenic bacteria expressing interleukin-10 in Crohn’s disease. Clin. Gastroenterol. Hepatol. 4, 754–759 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Nash, A. K. et al. The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome 5, 153 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Sokol, H. et al. Fungal microbiota dysbiosis in IBD. Gut 66, 1039–1048 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Enaud, R. et al. The mycobiome: a neglected component in the microbiota–gut–brain axis. Microorganisms 6, 22 (2018).

  49. Martins, F. S. et al. Oral treatment with Saccharomyces cerevisiae strain UFMG 905 modulates immune responses and interferes with signal pathways involved in the activation of inflammation in a murine model of typhoid fever. Int. J. Med. Microbiol. 301, 359–364 (2011).

    Article  PubMed  Google Scholar 

  50. Rizzetto, L. et al. Fungal chitin induces trained immunity in human monocytes during cross-talk of the host with Saccharomyces cerevisiae. J. Biol. Chem. 291, 7961–7972 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zanello, G. et al. Saccharomyces cerevisiae modulates immune gene expressions and inhibits ETEC-mediated ERK1/2 and p38 signaling pathways in intestinal epithelial cells. PLoS ONE 6, e18573 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chiaro, T. R. et al. A member of the gut mycobiota modulates host purine metabolism exacerbating colitis in mice. Sci. Transl. Med. 9, eaaf9044 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Rinaldi, M., Perricone, R., Blank, M., Perricone, C. & Shoenfeld, Y. Anti-Saccharomyces cerevisiae autoantibodies in autoimmune diseases: from bread baking to autoimmunity. Clin. Rev. Allergy Immunol. 45, 152–161 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Tiago, F. C. et al. Effect of Saccharomyces cerevisiae strain UFMG A-905 in experimental model of inflammatory bowel disease. Benef. Microbes 6, 807–815 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Hudson, L. E. et al. Functional heterologous protein expression by genetically engineered probiotic yeast Saccharomyces boulardii. PLoS ONE 9, e112660 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Durmusoglu, D. et al. In situ biomanufacturing of small molecules in the mammalian gut by probiotic Saccharomyces boulardii. ACS Synth. Biol. 10, 1039–1105 (2021).

  57. Takemura, N. et al. Eosinophil depletion suppresses radiation-induced small intestinal fibrosis. Sci. Transl. Med. 10, eaan0333 (2018).

    Article  PubMed  CAS  Google Scholar 

  58. Wilhelm, K. et al. Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R. Nat. Med. 16, 1434–1438 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Berer, K. et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Rothhammer, V. et al. Microglial control of astrocytes in response to microbial metabolites. Nature 557, 724–728 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Bashor, C. J., Helman, N. C., Yan, S. & Lim, W. A. Using engineered scaffold interactions to reshape MAP kinase pathway signaling dynamics. Science 319, 1539–1543 (2008).

    Article  CAS  PubMed  Google Scholar 

  62. Scott, B. M. et al. Coupling of human rhodopsin to a yeast signaling pathway enables characterization of mutations associated with retinal disease. Genetics 211, 597–615 (2019).

    Article  CAS  PubMed  Google Scholar 

  63. Rafehi, M. et al. Molecular recognition of agonists and antagonists by the nucleotide-activated G protein-coupled P2Y2 receptor. J. Med. Chem. 60, 8425–8440 (2017).

    Article  CAS  PubMed  Google Scholar 

  64. Wiederstein, M. & Sippl, M. J. ProSA-web: interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic Acids Res. 35, W407–W410 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Lovell, S. C. et al. Structure validation by Cα geometry: ϕ,ψ and Cβ deviation. Proteins 50, 437–450 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Lee, G. R. & Seok, C. Galaxy7TM: flexible GPCR–ligand docking by structure refinement. Nucleic Acids Res. 44, W502–W506 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ballesteros, J. A. & Weinstein, H. Integrated methods for the construction of three-dimensional models and computational probing of structure–function relations in G protein-coupled receptors. In Methods in Neurosciences 25 (ed. Stuart, C. S.), 366–428 (Academic Press, 1995).

  68. Ryan, O. W. et al. Selection of chromosomal DNA libraries using a multiplex CRISPR system. eLife 3, e03703 (2014).

    Article  PubMed Central  Google Scholar 

  69. Prykhozhij, S. V., Rajan, V., Gaston, D. & Berman, J. N. CRISPR multitargeter: a web tool to find common and unique CRISPR single guide RNA targets in a set of similar sequences. PLoS ONE 10, e0119372 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Pliatsika, V. & Rigoutsos, I. ‘Off-Spotter’: very fast and exhaustive enumeration of genomic lookalikes for designing CRISPR/Cas guide RNAs. Biol. Direct 10, 4 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Knowles, A. F. The GDA1_CD39 superfamily: NTPDases with diverse functions. Purinergic Signal. 7, 21–45 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Cox, L. M. et al. Calorie restriction slows age-related microbiota changes in an Alzheimer’s disease model in female mice. Sci. Rep. 9, 17904 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yoon, S. H. et al. Introducing EzBioCloud: a taxonomically united database of 16S rRNA gene sequences and whole-genome assemblies. Int. J. Syst. Evol. Microbiol. 67, 1613–1617 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Lozupone, C., Lladser, M. E., Knights, D., Stombaugh, J. & Knight, R. UniFrac: an effective distance metric for microbial community comparison. ISME J. 5, 169–172 (2011).

    Article  PubMed  Google Scholar 

  76. Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Erben, U. et al. A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int. J. Clin. Exp. Pathol. 7, 4557–4576 (2014).

    PubMed  PubMed Central  Google Scholar 

  78. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Babicki, S. et al. Heatmapper: web-enabled heat mapping for all. Nucleic Acids Res. 44, W147–W153 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants NS102807, ES02530, ES029136 and AI126880 from the NIH; RG4111A1 from the National MS Society; PA-1604-08459 from the International Progressive MS Alliance; and NSERC 492911 from the Natural Sciences Engineering Research Council of Canada. B.M.S. was supported by an Ontario Graduate Scholarship and by the Professional Research Experience Program agreement between the National Institute of Standards and Technology (NIST) and the University of Maryland. C.G.-V. was supported by an Alfonso Martín Escudero Foundation postdoctoral fellowship and by postdoctoral fellowship ALTF 610-2017 from the European Molecular Biology Organization. J.P. was supported by grant 2019/04780-7 from the São Paulo Research Foundation (FAPESP). This paper is dedicated to the memory of Dan S. Tawfik. We thank Z. Kelman and J. Marino of the University of Maryland Institute for Bioscience and Biotechnology Research for use of their incubators and W. Shaw of Imperial College London for providing yeast codon-optimized sfGFP. We thank the Harvard Medical School Rodent Histopathology Core, which provided histopathology service. We thank the NeuroTechnology Studio at Brigham and Women’s Hospital for providing access to the Leica DMi8 fluorescent microscope and consultation on data acquisition and data analysis. B.M.S. is an International Associate of the NIST. The NIST notes that certain commercial equipment, instruments and materials are identified in this paper to describe an experimental procedure as completely as possible. In no case does this identification imply a recommendation or endorsement by the NIST, nor does it imply that the materials, instruments or equipment are necessarily the best available for the purpose. The opinions expressed in this article are the authors’ own and do not necessarily represent the views of the NIST.

Author information

Authors and Affiliations

Authors

Contributions

B.M.S., C.G.-V., L.M.S., J.A.d.S.P., A.P., P.H., M.O’.B. and S.K.C. performed in vitro and in vivo experiments; Z.L., L.M.C. and C.G.-V. performed bioinformatic analysis; P.M.M.-V. discussed and interpreted findings; B.M.S., C.G.-V. and F.J.Q. wrote the manuscript with input from co-authors; and B.S.W.C., S.G.P. and F.J.Q. contributed equally to the design and supervision of the study and editing the manuscript.

Corresponding author

Correspondence to Francisco J. Quintana.

Ethics declarations

Competing interests

B.M.S., C.G.-V., B.S.W.C., S.G.P. and F.J.Q. filed a patent for the use of engineered yeast to treat inflammation. All other authors declare no competing interests.

Additional information

Peer review information Nature Medicine thanks Cathryn Nagler and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Saheli Sadanand was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Response to eATP over time of engineered mating pathway.

a,b, Yeasts from the BS016 strain transformed with plasmid pRS316 pTDH3 P2Y2 (WT human P2Y2 receptor) were incubated with 100 μM ATP in 300 μL (a) or 5 mL SC-URA media (b); and mCherry fluorescence was quantified. Data points represent the mean of 2 colonies. c, Engineered mating pathway response to UTP using the wild-type (WT) human P2Y2 receptor and various yeast strains with integrated Gpa1-Gα chimeras as follows: BS019 (G14), BS020 (Gq), BS016 (Gi3). Following incubation for 6h with the indicated UTP concentration, the activation of the mating pathway was monitored by quantifying mCherry fluorescence by flow cytometry. Data points represent the mean of 2 colonies.

Source data

Extended Data Fig. 2 Strategy for directed evolution of human P2Y2 receptor.

During each FACS sort the top ~1% of mCherry fluorescence was collected. ‘Recovered’ refers to the number of yeast colonies obtained after plating sorted cells on selective media.

Extended Data Fig. 3 Apyrase Genes and ATP concentration in yeast supernatants.

a, Sequence Alignment of Apyrase Genes. Human ENTPD1 (CD39), potato apyrase (RROP1) and wheat apyrase (TUAP1) were aligned using MUSCLE, in the MEGA6 alignment explorer. b, Measurement of a known ATP concentration in the presence of 5 μL yeast supernatant from strain CB008 (blue) or reaction buffer only (red). Yeast supernatant did not affect the measurement readout. n = 3 samples, error bars represent SD. c, To estimate the amount of active apyrase secreted by yeast, 50 μM ATP was incubated with the indicated concentration of commercial apyrase for 30 minutes at 30oC, with 5 μL supernatant from a culture of strain Ctrl, in a 50 μL reaction volume and residual ATP was quantified. No apyrase activity was observed when 31.3 pM commercial apyrase was added. n = 6 samples.

Source data

Extended Data Fig. 4 Engineered yeasts probiotics are viable in the mouse gut.

a, Colony forming units per mg of stool collected 6, 24 and 48h after oral gavage to the mice with either Ctrl KG, conAP KG or indAP KG yeast strains. n = 3 Ctrl, n = 5 conAP and n = 4 indAP samples per group. b, ATP relative levels in the specified portions of the gut of naïve and TNBS induced mice. n = 4 naïve, n = 6 TNBS mice per group. c, Gating strategy to measure mCherry positive yeasts in the fecal content. d, mCherry positive yeasts (% of total GFP yeast) measured by flow cytometry in the fecal content of the specified portion of the gut after 2h from oral gavage to naïve mice with ATP inducible strain indCherry KG. ATP levels were measured in the same portions of the gut. n = 3 samples for cytometry and n = 8 for eATP levels. e, mCherry positive yeasts (% of total GFP yeast) quantified by flow cytometry in the fecal content of the specified portion of the gut from TNBS treated mice 2 hours after oral gavage with indCherry KG or WTCherry KG yeast strains. f, Changes in body weight during the course of DSS-induced colitis in mice treated with engineered yeasts starting 3 days before DSS administration. n = 4 mice Healthy control and n = 3 otherwise. Two-way ANOVA followed by Tukey’s post-hoc test, ns = not significant.

Source data

Extended Data Fig. 5 Integration of P2Y2 mutants with CRISPR STAR Method. Plasmid pCAS AarI.

Custom multiple cloning site inserted at the XmaI and BglII sites in the pCAS plasmid, obtained from AddGene68. Image generated with CLC Sequence Viewer.

Supplementary information

Supplementary Information

Supplementary Tables 1–6 and Supplementary Figs. 1–3.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Scott, B.M., Gutiérrez-Vázquez, C., Sanmarco, L.M. et al. Self-tunable engineered yeast probiotics for the treatment of inflammatory bowel disease. Nat Med 27, 1212–1222 (2021). https://doi.org/10.1038/s41591-021-01390-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01390-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing