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:

A genetic system for Akkermansia muciniphila reveals a role for mucin foraging in gut colonization and host sterol biosynthesis gene expression

Abstract

Akkermansia muciniphila, a mucophilic member of the gut microbiota, protects its host against metabolic disorders. Because it is genetically intractable, the mechanisms underlying mucin metabolism, gut colonization and its impact on host physiology are not well understood. Here we developed and applied transposon mutagenesis to identify genes important for intestinal colonization and for the use of mucin. An analysis of transposon mutants indicated that de novo biosynthesis of amino acids was required for A. muciniphila growth on mucin medium and that many glycoside hydrolases are redundant. We observed that mucin degradation products accumulate in internal compartments within bacteria in a process that requires genes encoding pili and a periplasmic protein complex, which we term mucin utilization locus (MUL) genes. We determined that MUL genes were required for intestinal colonization in mice but only when competing with other microbes. In germ-free mice, MUL genes were required for A. muciniphila to repress genes important for cholesterol biosynthesis in the colon. Our genetic system for A. muciniphila provides an important tool with which to uncover molecular links between the metabolism of mucins, regulation of lipid homeostasis and potential probiotic activities.

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: A. muciniphila accumulates mucin glycans and requires amino acid biosynthesis for replication in mucin as a sole carbon and nitrogen source.
Fig. 2: The metabolic requirements for A. muciniphila to colonize the gastrointestinal tract increase as the host microbiota becomes more complex.
Fig. 3: MULs encode a mucin transport complex in A. muciniphila.
Fig. 4: Mucin utilization enables A. muciniphila to compete against members of the microbiota and leads to repression of genes in cholesterol biosynthesis.

Similar content being viewed by others

Data availability

Analysed data, including INSeq, RNA-seq and mass spectrometry outputs, as well as primer and adaptor sequences are available in Supplementary Data 1. Sequencing data used for this study can be found in the National Center for Biotechnology Information BioProject database with the accession code PRJNA955715. Unprocessed data from mass spectrometry and additional supporting data are available upon reasonable request from the corresponding authors. Source data are provided with this paper.

Code availability

Code for analysing the INSeq data is available at https://github.com/pmalkus/Akk_INseq_paper.

References

  1. Plovier, H. et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 23, 107–113 (2017).

    CAS  PubMed  Google Scholar 

  2. Blacher, E. et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 572, 474–480 (2019).

    CAS  PubMed  Google Scholar 

  3. Xie, J. et al. Akkermansia muciniphila protects mice against an emerging tick-borne viral pathogen. Nat. Microbiol. 8, 91–106 (2023).

    CAS  PubMed  Google Scholar 

  4. Derosa, L. et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat. Med. 28, 315–324 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Yoon, H. S. et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 6, 563–573 (2021).

    CAS  PubMed  Google Scholar 

  6. Zhang, Q. et al. Genetic mapping of microbial and host traits reveals production of immunomodulatory lipids by Akkermansia muciniphila in the murine gut. Nat. Microbiol. 8, 424–440 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Johansson, M. E. V., Larsson, J. M. H. & Hansson, G. C. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host–microbial interactions. Proc. Natl Acad. Sci. USA 108, 4659–4665 (2011).

    CAS  PubMed  Google Scholar 

  8. Wlodarska, M. et al. Indoleacrylic acid produced by commensal Peptostreptococcus species suppresses inflammation. Cell Host Microbe 22, 25–37 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Shon, D. J. et al. An enzymatic toolkit for selective proteolysis, detection, and visualization of mucin-domain glycoproteins. Proc. Natl Acad. Sci. USA 117, 21299–21307 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Trastoy, B., Naegeli, A., Anso, I., Sjögren, J. & Guerin, M. E. Structural basis of mammalian mucin processing by the human gut O-glycopeptidase OgpA from Akkermansia muciniphila. Nat. Commun. 11, 4844 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Medley, B. J. et al. A previously uncharacterized O-glycopeptidase from Akkermansia muciniphila requires the Tn-antigen for cleavage of the peptide bond. J. Biol. Chem. 298, 102439 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Crouch, L. I. et al. Prominent members of the human gut microbiota express endo-acting O-glycanases to initiate mucin breakdown. Nat. Commun. 11, 4017 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Meng, X. et al. A purified aspartic protease from Akkermansia muciniphila plays an important role in degrading Muc2. Int. J. Mol. Sci. 21, 72 (2020).

    CAS  Google Scholar 

  14. Xu, W., Yang, W., Wang, Y., Wang, M. & Zhang, M. Structural and biochemical analyses of β-N-acetylhexosaminidase Am0868 from Akkermansia muciniphila involved in mucin degradation. Biochem. Biophys. Res. Commun. 529, 876–881 (2020).

    CAS  PubMed  Google Scholar 

  15. Kosciow, K. & Deppenmeier, U. Characterization of a phospholipid-regulated β-galactosidase from Akkermansia muciniphila involved in mucin degradation. MicrobiologyOpen 8, e00796 (2019).

    PubMed  PubMed Central  Google Scholar 

  16. Guo, B.-S. et al. Cloning, purification and biochemical characterisation of a GH35 beta-1,3/beta-1,6-galactosidase from the mucin-degrading gut bacterium Akkermansia muciniphila. Glycoconj. J. 35, 255–263 (2018).

    CAS  PubMed  Google Scholar 

  17. Chen, X. et al. Crystallographic evidence for substrate-assisted catalysis of β-N-acetylhexosaminidas from Akkermansia muciniphila. Biochem. Biophys. Res. Commun. 511, 833–839 (2019).

    CAS  PubMed  Google Scholar 

  18. Pruss, K. M. et al. Mucin-derived O-glycans supplemented to diet mitigate diverse microbiota perturbations. ISME J. 15, 577–591 (2021).

    CAS  PubMed  Google Scholar 

  19. Goodman, A. L., Wu, M. & Gordon, J. I. Identifying microbial fitness determinants by insertion sequencing using genome-wide transposon mutant libraries. Nat. Protoc. 6, 1969–1980 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Anzai, I. A., Shaket, L., Adesina, O., Baym, M. & Barstow, B. Rapid curation of gene disruption collections using Knockout Sudoku. Nat. Protoc. 12, 2110–2137 (2017).

    CAS  PubMed  Google Scholar 

  21. Derrien, M., Vaughan, E. E., Plugge, C. M. & de Vos, W. M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 54, 1469–1476 (2004).

    CAS  PubMed  Google Scholar 

  22. Hansson, G. C. Mucins and the microbiome. Annu. Rev. Biochem. 89, 769–793 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lensmire, J. M. & Hammer, N. D. Nutrient sulfur acquisition strategies employed by bacterial pathogens. Curr. Opin. Microbiol. 47, 52–58 (2019).

    CAS  PubMed  Google Scholar 

  24. Lombard, V., Golaconda Ramulu, H., Drula, E., Coutinho, P. M. & Henrissat, B. The carbohydrate-active enzymes database (CAZy) in 2013. Nucleic Acids Res. 42, 490–495 (2014).

    Google Scholar 

  25. Kostopoulos, I. et al. Akkermansia muciniphila uses human milk oligosaccharides to thrive in the early life conditions in vitro. Sci. Rep. 10, 14330 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Ottman, N. et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 12, e0173004 (2017).

    PubMed  PubMed Central  Google Scholar 

  27. Van Passel, M. W. J. et al. The genome of Akkermansia muciniphila, a dedicated intestinal mucin degrader, and its use in exploring intestinal metagenomes. PLoS ONE 6, e16876 (2011).

    PubMed  PubMed Central  Google Scholar 

  28. Ottman, N. et al. Genome-scale model and omics analysis of metabolic capacities of Akkermansia muciniphila reveal a preferential mucin-degrading lifestyle. Appl. Environ. Microbiol. 83, e01014–e01017 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Thibault, D. et al. Droplet Tn-Seq combines microfluidics with Tn-Seq for identifying complex single-cell phenotypes. Nat. Commun. 10, 5729 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Ottman, N. et al. Characterization of outer membrane proteome of Akkermansia muciniphila reveals sets of novel proteins exposed to the human intestine. Front. Microbiol. 7, 1157 (2016).

    PubMed  PubMed Central  Google Scholar 

  31. Schwalm, N. D. & Groisman, E. A. Navigating the gut buffet: control of polysaccharide utilization in Bacteroides spp. Trends Microbiol. 25, 1005–1015 (2017).

    CAS  PubMed  Google Scholar 

  32. Mistry, J. et al. Pfam: the protein families database in 2021. Nucleic Acids Res. 49, D412–D419 (2021).

    CAS  PubMed  Google Scholar 

  33. Marchler-Bauer, A. et al. CDD/SPARCLE: functional classification of proteins via subfamily domain architectures. Nucleic Acids Res. 45, D200–D203 (2017).

    CAS  PubMed  Google Scholar 

  34. Cortajarena, A. L. & Regan, L. Ligand binding by TPR domains. Protein Sci. 15, 1193–1198 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Xiang, R., Wang, J., Xu, W., Zhang, M. & Wang, M. Amuc_1102 from Akkermansia muciniphila adopts an immunoglobulin-like fold related to archaeal type IV pilus. Biochem. Biophys. Res. Commun. 547, 59–64 (2021).

    CAS  PubMed  Google Scholar 

  36. Mou, L. et al. Crystal structure of monomeric Amuc-1100 from Akkermansia muciniphila. Acta Crystallogr. F Struct. Biol. Commun. 76, 168–174 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Velcich, A. et al. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science 295, 1726–1729 (2002).

    CAS  PubMed  Google Scholar 

  38. Becken, B. et al. Genotypic and phenotypic diversity among human isolates of Akkermansia muciniphila. mBio 12, e00478-21 (2021).

    PubMed  PubMed Central  Google Scholar 

  39. Roux, D. et al. Identification of poly-N-acetylglucosamine as a major polysaccharide component of the Bacillus subtilis biofilm matrix. J. Biol. Chem. 290, 19261–19272 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Glenwright, A. J. et al. Structural basis for nutrient acquisition by dominant members of the human gut microbiota. Nature 541, 407–411 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Bolam, D. N. & van den Berg, B. TonB-dependent transport by the gut microbiota: novel aspects of an old problem. Curr. Opin. Struct. Biol. 51, 35–43 (2018).

    CAS  PubMed  Google Scholar 

  42. Boedeker, C. et al. Determining the bacterial cell biology of Planctomycetes. Nat. Commun. 8, 14853 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Holden, H. M., Rayment, I. & Thoden, J. B. Structure and function of enzymes of the Leloir pathway for galactose metabolism. J. Biol. Chem. 278, 43885–43888 (2003).

    CAS  PubMed  Google Scholar 

  44. Faham, S. et al. The crystal structure of a sodium galactose transporter reveals mechanistic insights into Na+/sugar symport. Science 321, 810–814 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Depommier, C. et al. Pasteurized Akkermansia muciniphila increases whole-body energy expenditure and fecal energy excretion in diet-induced obese mice. Gut Microbes 11, 1231–1245 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Lukovac, S. et al. Differential modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of host peripheral lipid metabolism and histone acetylation in mouse gut organoids. mBio 5, e01438–14 (2014).

    PubMed  PubMed Central  Google Scholar 

  47. Schaum, N. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    PubMed Central  Google Scholar 

  48. Wang, B. et al. Phospholipid remodeling and cholesterol availability regulate intestinal stemness and tumorigenesis. Cell Stem Cell 22, 206–220 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. McFarlane, M. R. et al. Scap is required for sterol synthesis and crypt growth in intestinal mucosa. J. Lipid Res. 56, 1560–1571 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Depommier, C. et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat. Med. 25, 1096–1103 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Goodman, A. L. et al. Identifying genetic determinants needed to establish a human gut symbiont in its habitat. Cell Host Microbe 6, 279–289 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Grondin, J. M., Tamura, K., Déjean, G., Abbott, D. W. & Brumer, H. Polysaccharide utilization loci: fueling microbial communities. J. Bacteriol. 199, e00860-16 (2017).

    PubMed  PubMed Central  Google Scholar 

  53. Arnosti, C. Fluorescent derivatization of polysaccharides and carbohydrate-containing biopolymers for measurement of enzyme activities in complex media. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 793, 181–191 (2003).

    CAS  Google Scholar 

  54. Pan, Y. & Kaatz, L. Use of image-based flow cytometry in bacterial viability analysis using fluorescent probes. Curr. Protoc. Microbiol. 27, 2C.5.1–2C.5.11 (2012).

    Google Scholar 

  55. Aronesty, E. Comparison of sequencing utility programs. Open Bioinform. J. 7, 1–8 (2013).

    Google Scholar 

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

    CAS  PubMed  Google Scholar 

  57. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  58. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS J. Integr. Biol. 16, 284–287 (2012).

    CAS  Google Scholar 

  59. Wolk, C. P. et al. Paired cloning vectors for complementation of mutations in the cyanobacterium Anabaena sp. strain PCC 7120. Arch. Microbiol. 188, 551–563 (2007).

    CAS  PubMed  Google Scholar 

  60. Stothard, P. The sequence manipulation suite: JavaScript programs for analyzing and formatting protein and DNA sequences. BioTechniques 28, 1102–1104 (2000).

    CAS  PubMed  Google Scholar 

  61. O’Toole, G. A. et al. Genetic approaches to study of biofilms. Methods Enzymol. 310, 91–109 (1999).

    PubMed  Google Scholar 

  62. Najah, M., Griffiths, A. D. & Ryckelynck, M. Teaching single-cell digital analysis using droplet-based microfluidics. Anal. Chem. 84, 1202–1209 (2012).

    CAS  PubMed  Google Scholar 

  63. Shames, S. R. et al. Multiple Legionella pneumophila effector virulence phenotypes revealed through high-throughput analysis of targeted mutant libraries. Proc. Natl Acad. Sci. USA 114, E10446–E10454 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Pritchard, J. R. et al. ARTIST: high-resolution genome-wide assessment of fitness using transposon-insertion sequencing. PLoS Genet. 10, e1004782 (2014).

    PubMed  PubMed Central  Google Scholar 

  65. DeJesus, M. A., Ambadipudi, C., Baker, R., Sassetti, C. & Ioerger, T. R. TRANSIT—a software tool for Himar1 TnSeq analysis. PLoS Comput. Biol. 11, e1004401 (2015).

    PubMed  PubMed Central  Google Scholar 

  66. Karp, P. D. et al. The BioCyc collection of microbial genomes and metabolic pathways. Brief. Bioinformatics 20, 1085–1093 (2018).

    Google Scholar 

  67. Ansaldo, E. et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science 364, 1179–1184 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Collado, M. C., Derrien, M., Isolauri, E., De Vos, W. M. & Salminen, S. Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl. Environ. Microbiol. 73, 7767–7770 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    PubMed  PubMed Central  Google Scholar 

  70. Petersen, T. N., Brunak, S., von Heijne, G. & Nielsen, H. SignalP 4.0: discriminating signal peptides from transmembrane regions. Nat. Methods 8, 785–786 (2011).

    CAS  PubMed  Google Scholar 

  71. Shannon, P. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Thompson, L. R. et al. A communal catalogue reveals Earth’s multiscale microbial diversity. Nature 551, 457–463 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  74. McMurdie, P. J. & Holmes, S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  76. Paley, S. et al. The omics dashboard for interactive exploration of gene-expression data. Nucleic Acids Res. 45, 12113–12124 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are thankful to O. Kuddar and E. Rivas for support with the assembly of arrayed Tn mutant libraries; J. Granek for the base trimming code; A. Sharma for preparing sequencing libraries; and members of the R.H.V. laboratory for critical reading of the manuscript. We thank L. Augenlicht at the Albert Einstein College of Medicine for providing the Muc2−/− mice. This work was supported by National Institutes of Health awards AI142376 and DK110496 (to R.H.V.), American Heart Association award 18POST34070017 (to L.E.D.) and a fellowship from the Natural Sciences and Engineering Research Council of Canada (PDF4878642016 to L.E.D.).

Author information

Authors and Affiliations

Authors

Contributions

R.H.V., L.E.D. and P.N.M. designed the research. L.E.D., P.N.M., M.V., L.D. and E.A. performed the experiments and analysed the data. L.E.D. prepared the figures. P.M.N. wrote the INSeq analysis code. Z.C.H. and J.L. contributed to running the SCFA analysis. M.V. performed the microfluidic droplet experiments. L.D. performed the live imaging experiments. L.E.D. and R.H.V. wrote the manuscript. R.H.V. supervised the project. All authors reviewed and edited the manuscript.

Corresponding authors

Correspondence to Lauren E. Davey or Raphael H. Valdivia.

Ethics declarations

Competing interests

R.H.V. is a founder of Bloom Science.

Peer review

Peer review information

Nature Microbiology thanks Matthew Waldor and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Akkermansia sp. are mucin specialists and the acquisition of mucin by A. muciniphila is selective and energy dependent.

(a) Growth curves, as assessed by optical density (OD600) of a range of Gram-positive and Gram-negative mucin-degrading intestinal microbes, including A. muciniphila and A. glycaniphila, in the indicated medium. (b) A. muciniphila and Bacteroides thetaiotaomicron grown with fluorescein-mucin. The cells were grown with fluorescein mucin in a modified version of synthetic media with 0.25% mucin as the sole carbon source. Membranes were labelled with FM4-64. Experiments were repeated twice. (c-d) Mucin uptake is a specific and active process. A. muciniphila grown in the presence of either fluorescein-mucin or fluorescein-dextran (green) for 3 h and stained with anti-Akkermansia anti-sera (anti-Akk). All microscopy was performed at least three times (c). Flow cytometric analysis of cells grown in the presence of fluorescein-mucin for 3 h, with or without pre-treatment with CCCP. Cells for flow cytometry were gated for the anti-Akkermansia positive population and the numbers under each curve represent the mean fluorescent intensity of fluorescein-mucin (d). A. muciniphila grown with fluorescein-mucin for 3 h without CCCP or with CCCP treatment (e). Flow cytometry analyses of A. muciniphila grown with the cell permanent esterase carboxyfluorescein diacetate (CFDA) in the presence and absence of CCCP, and after heat inactivation (f). Scale bar, 1 μm. Error bars represent the standard error of the mean.

Source data

Extended Data Fig. 2 Transposon mutagenesis in A. muciniphila.

(a) Map of the A. muciniphila optimized INSeq plasmid. (b) Overview of the A. muciniphila conjugation protocol. (c) PCR analysis confirming transposition. DNA from representative Tn mutants was amplified with primers for A. muciniphila specific 16 S rRNA, the bla gene located on the delivery plasmid backbone, and the cat gene located with the transposon. This analysis was performed for every transposition experiment. (d) Southern blot analysis of Tn mutant DNA digested with HindIII and probed with DIG-labelled probes that recognize the cat gene in the Tn insert. Data is representative of two experiments with similar results. (e-f) A Cartesian mapping strategy to identify Tn insertions. (e) Trade-off between genome coverage and clonal redundancy, using simulated subsets of the arrayed collection optimized for low redundancy. A series of 96-well plates drawn from the arrayed collection that minimizes clonal redundancy was identified by simulation. The tradeoff between increasing genome coverage (orange) and increasing clonal redundancy (blue) as the number of plates included from the optimized series grows (X-axis). (f) Estimating location mapping accuracy for different sizes of the optimized library. The distribution of clonal replicates for increasing sizes of the optimized library was drawn from the simulation. The estimated number of clones present in one, two, three, and four replicates are shown as a function of increasing collection size. For orthogonal pooling and Cartesian location mapping the search space for an individual clone scales as the number of replicates to the power of the number of pooling dimensions. A clone present in only one well has a unique plate-well address (1^3), while a clone with present three times in the collection would be mapped to 27 potential Plate-Row-Col locations (3^3).

Source data

Extended Data Fig. 3 INSeq analysis of relative nutritional requirements for A. muciniphila to grow in mucin medium and the role of putative glycan hydrolases.

Plot of INSeq data from Tn mutant pools grown for eight generations in mucin medium where each dot represents all inserts in a specific gene. Genes that belong to KEGG amino acid biosynthesis pathways are highlighted for cultures grown in (a) mucin medium and (b) mucin medium supplemented with Phytone. Predicted glycosyl hydrolases for A. muciniphila BAA-835 were identified using the CAZy database and highlighted on the INSeq plot for cultures grown in (c) mucin and in (d) mucin medium with Phytone. Statistical analysis on INSeq data was performed with a Mann-Whitney Utest. (e) Droplet-seq analysis of A. muciniphila grown in mucin medium microdroplets. Tn mutants (Arrayed Pool) were injected into a microfluidic device at a low density to generate on average less than one bacterium per droplet. The graph displays the INSeq analysis and Log2 fold change for cultures grown in mucin in batch culture (8 generations) versus single cell growth in droplets (72 h). Selected genes that were depleted in one condition relative to the other are highlighted on the plot. GH, glycosyl hydrolase.

Source data

Extended Data Fig. 4 A significant proportion of A. muciniphila genes required for growth in mucin medium are specific to Akkermansia/Verrucomicrobia.

(a) Number of genes required for optimal A. muciniphila growth in mucin medium that lack functional annotations. Genes corresponding to Tn mutants with a Log2 > 2 fold decrease in abundance in mucin medium were used as the query for a BLAST search to identify potential homologs. The plot represents the number of genes encoding hypothetical proteins that were unique to Akkermansia spp. (Akk), homologs in other members of the PVC super phylum (PVC), homologs in other bacteria (other), and genes annotated as conserved hypothetical proteins (conserved). (b) Distribution of genes with Pfam designations belonging to pili or type II secretion families (Pili/T2SS), or TPR families in the INSeq analysis of genes required for growth in mucin medium in vitro, (c) in the cecum of germ-free mice, and (d) in the cecum of conventional mice.

Source data

Extended Data Fig. 5 Evidence for the presence of a stable Mul1A-Mul1B protein complex.

Transcriptional analysis of Mul1 operons. (a) View of RNA-seq reads generated from wild type A. muciniphila grown in mucin medium mapped to genes in the mul1 and mul2 loci. (b) Growth curves for wild type A. muciniphila and mutants in mul1B and mul2B grown in triplicate in synthetic medium or with mucin as the sole carbon and nitrogen source and corresponding microscopy with FL-mucin (green). Cells are stained with anti-Akkermansia antisera (white). The scale bar is 1 μm. (c) Coomassie blue stained SDS-PAGE gel showing eluted proteins following immunoprecipitation with anti-Mul1 antibodies. Immunoprecipitations were performed with cell lysates from wild type A. muciniphila and in mul1A mutants. (d) Depiction of Conserved Domains (colours) in Muc5AC and locations of peptides identified as co-precipitating with Mul1A (vertical bars). The experiment was performed in triplicate.

Source data

Extended Data Fig. 6 Mucin utilization is required for A. muciniphila to compete in CONV mice and in Muc2-/- mice.

A breeding colony of Akkermansia-free mice (Akk-free) was generated to facilitate mouse colonization without antibiotic pre-treatment. (a-c) Comparison of the microbiota of Akkermansia colonized (Akk-colonized) and Akk-free mice by 16 S rRNA gene sequencing. (a) Relative abundances of fecal bacteria at the genus level in Akk-colonized and Akk-free mice. Each sample was obtained from separately housed mice (n = 3 per group). (b) Principal Coordinates Analysis (PCoA) performed on weighted UniFrac distances. Statistical significance was determined by Permutational Multivariate Analysis of Variance (PERMANOVA). (c) Relative abundances of potential mucin-degrading taxa at the family level. The centre line is the mean, and the whiskers show the minimum and maximum. (d) Linear discriminant analysis Effect Size (LEfSe) analysis of Akk-colonized and Akk-free mice. The Kruskal-Wallis test was used to detect features with a significant differential abundance (p < 0.05). (e) CONV mice were pre-treated with antibiotics and gavaged with a 1:1 mix of WT and mutant A. muciniphila prepared with a fecal slurry from Akk-free mice to partially reconstitute the microbiota (n = 6 per group). Bacterial loads in fecal pellets were quantified by qPCR, each point represents one cage. (f) Colonization of mucin deficient Muc2-/- mice with A. muciniphila. Each point represents the average A. muciniphila per gram of feces (WT, n = 4; mul1A::Tn, n = 4; mul2A::Tn, n = 6). (g) Competition between wild type A. muciniphila and the mul1A::Tn mutant in Muc2-/- mice. Mice were gavaged with a 1:1 mix of wild type and mutant and abundance was monitored over time using strain specific primers. Each point represents the average amount of A. muciniphila (n = 4), error bars represent the standard error.

Source data

Extended Data Fig. 7 The impact of mucin utilization by A. muciniphila in colonization along the GI tract, SCFA production and transcriptional responses.

(a) Abundance of A. muciniphila wild type and mul1A mutants along the GI tract of female GF mice (n = 3). Intestinal contents were scraped from sections along the GI tract and A. muciniphila levels were quantified by qPCR. Data are presented as mean values +/- SEM. The analysis was carried out with the same female mice that were used for RNAseq. (b) Expression of cholesterol biosynthesis genes in male and female mice, and control mice gavaged with sterile PBS. (c) Normalized expression of genes that are pivotal to cholesterol biosynthesis (Hmgcr) and uptake (Ldlr) in relation to cecal acetate and propionate levels. (d) Representative single cell RNAseq expression data from the Tabula Muris47. Violin plots show the expression of Ldlr and Hmgcr in mouse colonic epithelial and goblet cells.

Source data

Supplementary information

Reporting Summary

Supplementary Video 1

View through an orthogonal section of a 3D reconstructed STED image of a mucinosome inside A. muciniphila. Fluorescein-labelled mucin is shown in magenta and the A. muciniphila cell surface (anti-Akk) is cyan.

Supplementary Video 2

Live imaging of A. muciniphila grown with fluorescein-labelled mucin under an agarose pad, with and without the addition of 50 μM CCCP. Images were captured every 30 s for 20 min. The arrows indicate cells with active mucinosome formation (untreated) and locations where mucin accumulates at the cell surface but fails to form mucinosomes in the presence of CCCP.

Supplementary Data 1

Data tables containing INSeq, RNA-seq and mass spectrometry outputs and primer and adaptor sequences.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unmodified gel and blot.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unmodified gel image.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

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

Davey, L.E., Malkus, P.N., Villa, M. et al. A genetic system for Akkermansia muciniphila reveals a role for mucin foraging in gut colonization and host sterol biosynthesis gene expression. Nat Microbiol 8, 1450–1467 (2023). https://doi.org/10.1038/s41564-023-01407-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-023-01407-w

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