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:

An alternative broad-specificity pathway for glycan breakdown in bacteria

Abstract

The vast majority of glycosidases characterized to date follow one of the variations of the ‘Koshland’ mechanisms1 to hydrolyse glycosidic bonds through substitution reactions. Here we describe a large-scale screen of a human gut microbiome metagenomic library using an assay that selectively identifies non-Koshland glycosidase activities2. Using this, we identify a cluster of enzymes with extremely broad substrate specificities and thoroughly characterize these, mechanistically and structurally. These enzymes not only break glycosidic linkages of both α and β stereochemistry and multiple connectivities, but also cleave substrates that are not hydrolysed by standard glycosidases. These include thioglycosides, such as the glucosinolates from plants, and pseudoglycosidic bonds of pharmaceuticals such as acarbose. This is achieved through a distinct mechanism of hydrolysis that involves oxidation/reduction and elimination/hydration steps, each catalysed by enzyme modules that are in many cases interchangeable between organisms and substrate classes. Homologues of these enzymes occur in both Gram-positive and Gram-negative bacteria associated with the gut microbiome and other body parts, as well as other environments, such as soil and sea. Such alternative step-wise mechanisms appear to constitute largely unrecognized but abundant pathways for glycan degradation as part of the metabolism of carbohydrates in bacteria.

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: Koshland glycosidases and non-Koshland thioglycoside substrates.
Fig. 2: Substrate structures and screening results.
Fig. 3: Genes in the P1C11 fosmid and B. thetaiotaomicron.
Fig. 4: Structures of the enzymes and the reactions that they catalyse.
Fig. 5: Genes in the P2B11 fosmid and structural information.
Fig. 6: The overall proposed mechanism of broad-specificity stepwise hydrolysis.

Similar content being viewed by others

Data availability

Data supporting the findings of this study are available within the Article and the Supplementary Information. All sequencing data are available at the end of the Supplementary Information and also from GenBank database with accession numbers PP693324 and PP693325. Structural models are deposited at the Protein Data Bank under accession codes 8TCD, 8TCR, 8TCS, 8TCT, 8TDA, 8TDE, 8TDF, 8TDH, 8TDI and 8V31Source data are provided with this paper.

References

  1. Koshland, D. E. Stereochemistry and the mechanism of enzymatic reactions. Biol. Rev. 28, 416–436 (1953).

    CAS  Google Scholar 

  2. Nasseri, S. A., Betschart, L., Opaleva, D., Rahfeld, P. & Withers, S. G. A mechanism-based approach to screening metagenomic libraries for discovery of unconventional glycosidases. Angew. Chem. 130, 11529–11534 (2018).

    ADS  Google Scholar 

  3. Wolfenden, R., Lu, X. & Young, G. Spontaneous hydrolysis of glycosides. J. Am. Chem. Soc. 120, 6814–6815 (1998).

    CAS  Google Scholar 

  4. Watts, A. G. et al. Trypanosoma cruzi trans-sialidase operates through a covalent sialyl-enzyme intermediate: tyrosine is the catalytic nucleophile. J. Am. Chem. Soc. 125, 7532–7533 (2003).

    CAS  PubMed  Google Scholar 

  5. Vocadlo, D. J. & Withers, S. G. Detailed comparative analysis of the catalytic mechanisms of β-N-acetylglucosaminidases from families 3 and 20 of glycoside hydrolases. Biochemistry 44, 12809–12818 (2005).

    CAS  PubMed  Google Scholar 

  6. Namchuk, M. N. & Withers, S. G. Mechanism of Agrobacterium β-glucosidase: kinetic analysis of the role of noncovalent enzyme/substrate interactions. Biochemistry 34, 16194–16202 (1995).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  8. Drula, E. et al. The carbohydrate-active enzyme database: functions and literature. Nucleic Acids Res. 50, D571–D577 (2022).

    CAS  PubMed  Google Scholar 

  9. Yip, V. L. Y. & Withers, S. G. Family 4 glycosidases carry out efficient hydrolysis of thioglycosides by an α,β-elimination mechanism. Angew. Chem. Int. Ed. 45, 6179–6182 (2006).

    CAS  Google Scholar 

  10. Jongkees, S. A. K. & Withers, S. G. Glycoside cleavage by a new mechanism in unsaturated glucuronyl hydrolases. J. Am. Chem. Soc. 133, 19334–19337 (2011).

    CAS  PubMed  Google Scholar 

  11. Rahfeld, P. et al. An enzymatic pathway in the human gut microbiome that converts A to universal O type blood. Nat. Microbiol. 4, 1475–1485 (2019).

    CAS  PubMed  Google Scholar 

  12. Lorenz, P. & Eck, J. Metagenomics and industrial applications. Nat. Rev. Microbiol. 3, 510–516 (2005).

    CAS  PubMed  Google Scholar 

  13. Viborg, A. H. et al. A subfamily roadmap of the evolutionarily diverse glycoside hydrolase family 16 (GH16). J. Biol. Chem. 294, 15973–15986 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Hallgren, J. et al. DeepTMHMM predicts alpha and beta transmembrane proteins using deep neural networks. Preprint at bioRxiv https://doi.org/10.1101/2022.04.08.487609 (2022).

  15. Teufel, F. et al. SignalP 6.0 predicts all five types of signal peptides using protein language models. Nat. Biotechnol. 40, 1023–1025 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Liou, C. S. et al. A metabolic pathway for activation of dietary glucosinolates by a human gut symbiont. Cell 180, 717–728 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Ravcheev, D. A., Godzik, A., Osterman, A. L. & Rodionov, D. A. Polysaccharides utilization in human gut bacterium Bacteroides thetaiotaomicron: comparative genomics reconstruction of metabolic and regulatory networks. BMC Genomics 14, 873 (2013).

    PubMed  PubMed Central  Google Scholar 

  18. Liu, H. et al. Functional genetics of human gut commensal Bacteroides thetaiotaomicron reveals metabolic requirements for growth across environments. Cell Rep. 34, 108789 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Yip, V. L. Y. et al. An unusual mechanism of glycoside hydrolysis involving redox and elimination steps by a family 4 β-glycosidase from Thermotoga maritima. J. Am. Chem. Soc. 126, 8354–8355 (2004).

    CAS  PubMed  Google Scholar 

  20. Jäger, M., Hartmann, M., de Vries, J. G. & Minnaard, A. J. Catalytic regioselective oxidation of glycosides. Angew. Chemie Int. Ed. 52, 7809–7812 (2013).

    Google Scholar 

  21. Morris, P. E., Hope, K. D. & Kiely, D. E. The isomeric composition of d-ribo-hexos-3-ulose (3-keto-d-glucose) in aqueous solution. J. Carbohydr. Chem. 8, 515–530 (1989).

    CAS  Google Scholar 

  22. Allouch, J. et al. The three-dimensional structures of two β-agarases. J. Biol. Chem. 278, 47171–47180 (2003).

    CAS  PubMed  Google Scholar 

  23. Lemke, A., Kiderlen, A. F. & Kayser, O. Amphotericin B. Appl. Microbiol. Biotechnol. 68, 151–162 (2005).

    CAS  PubMed  Google Scholar 

  24. Arsic, B. et al. 16-membered macrolide antibiotics: a review. Int. J. Antimicrob. Agents 51, 283–298 (2018).

    CAS  PubMed  Google Scholar 

  25. Zhao, Q. et al. Neriifolin from seeds of Cerbera manghas L. induces cell cycle arrest and apoptosis in human hepatocellular carcinoma HepG2 cells. Fitoterapia 82, 735–741 (2011).

    CAS  PubMed  Google Scholar 

  26. Sugawara, K. et al. Elsamicins A and B, new antitumor antibiotics related to chartreusin. 2. Structures of elsamicins A and B. J. Org. Chem. 52, 996–1001 (1987).

    CAS  Google Scholar 

  27. Martens, E. C. et al. Recognition and degradation of plant cell wall polysaccharides by two human gut symbionts. PLoS Biol. 9, e1001221 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. van Beeumen, J. & de Ley, J. Hexopyranoside: cytochrome c oxidoreductase from Agrobacterium tumefaciens. Eur. J. Biochem. 6, 331–343 (1968).

    PubMed  Google Scholar 

  29. Takeuchi, M., Asano, N., Kameda, Y. & Matsui, K. Physiological role of glucoside 3-dehydrogenase and cytochrome c551 in the sugar oxidizing system of Flavobacterium saccharophilum. J. Biochem. 103, 938–943 (1988).

    CAS  PubMed  Google Scholar 

  30. Bernaerts, M. J. & De Ley, J. Microbiological formation and preparation of 3-ketoglycosides from disaccharides. J. Gen. Microbiol. 22, 129–136 (1960).

    CAS  PubMed  Google Scholar 

  31. Grebner, E. E. & Feingold, D. S. d-Aldohexopyranoside dehydrogenase of Agrobacterium tumefaciens. Biochem. Biophys. Res. Commun. 19, 37–42 (1965).

    CAS  Google Scholar 

  32. Hayano, K. & Fukui, S. Purification and properties of 3-ketosucrose-forming enzyme from the cells of Agrobacterium tumefaciens. J. Biol. Chem. 242, 3655–3672 (1967).

    CAS  PubMed  Google Scholar 

  33. Kuritani, Y. et al. Conversion of levoglucosan into glucose by the coordination of four enzymes through oxidation, elimination, hydration, and reduction. Sci. Rep. 10, 20066 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Kaur, A. et al. Identification of levoglucosan degradation pathways in bacteria and sequence similarity network analysis. Arch. Microbiol. 205, 155 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Braune, A., Engst, W. & Blaut, M. Identification and functional expression of genes encoding flavonoid O- and C-glycosidases in intestinal bacteria. Environ. Microbiol. 18, 2117–2129 (2016).

    CAS  PubMed  Google Scholar 

  36. He, P. et al. Structural mechanism of a dual-functional enzyme DgpA/B/C as both a C-glycoside cleaving enzyme and an O- to C-glycoside isomerase. Acta Pharm. Sin. B 13, 246–255 (2023).

    CAS  PubMed  Google Scholar 

  37. Mori, T. et al. C-Glycoside metabolism in the gut and in nature: Identification, characterization, structural analyses and distribution of C-C bond-cleaving enzymes. Nat. Commun. 12, 6294 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zheng, S. et al. A newly isolated human intestinal bacterium strain capable of deglycosylating flavone C-glycosides and its functional properties. Microb. Cell Fact. 18, 94 (2019).

    PubMed  PubMed Central  Google Scholar 

  39. Taborda, A. et al. Mechanistic insights into glycoside 3-oxidases involved in C-glycoside metabolism in soil microorganisms. Nat. Commun. 14, 7289 (2023).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu, Q. P. et al. Bacterial glycosidases for the production of universal red blood cells. Nat. Biotechnol. 25, 454–464 (2007).

    CAS  PubMed  Google Scholar 

  41. Bell, A. et al. Uncovering a novel molecular mechanism for scavenging sialic acids in bacteria. J. Biol. Chem. 295, 13724–13736 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kaur, A. et al. Widespread family of NAD-dependent sulfoquinovosidases at the gateway to sulfoquinovose catabolism. J. Am. Chem. Soc. 145, 28216–28223 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bitter, J. et al. Enzymatic β-elimination in natural product O- and C-glycoside deglycosylation. Nat. Commun. 14, 7123 (2023).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Armstrong, Z., Rahfeld, P. & Withers, S. G. Discovery of new glycosidases from metagenomic libraries. Methods Enzymol. 597, 3–23 (2017).

  45. Chen, H.-M. et al. Synthesis and evaluation of sensitive coumarin-based fluorogenic substrates for discovery of α-N-acetyl galactosaminidases through droplet-based screening. Org. Biomol. Chem. 19, 789–793 (2021).

    ADS  CAS  PubMed  Google Scholar 

  46. Klock, H. E., Koesema, E. J., Knuth, M. W. & Lesley, S. A. Combining the polymerase incomplete primer extension method for cloning and mutagenesis with microscreening to accelerate structural genomics efforts. Proteins Struct. Funct. Genet. 71, 982–994 (2008).

    CAS  PubMed  Google Scholar 

  47. Engler, C., Kandzia, R. & Marillonnet, S. A one pot, one step, precision cloning method with high throughput capability. PLoS ONE 3, e3647 (2008).

    ADS  PubMed  PubMed Central  Google Scholar 

  48. Studier, F. W. Protein production by auto-induction in high density shaking cultures. Protein Expr. Purif. 41, 207–234 (2005).

    CAS  PubMed  Google Scholar 

  49. Barnhart, J. L. & Berk, R. N. Influence of paramagnetic ions and pH on proton NMR relaxation of biologic fluids. Invest. Radiol. 21, 132–136 (1986).

    CAS  PubMed  Google Scholar 

  50. Winter, G. xia2: an expert system for macromolecular crystallography data reduction. J. Appl. Crystallogr. 43, 186–190 (2010).

    ADS  CAS  Google Scholar 

  51. Kabsch, W. XDS. Acta Crystallogr. D 66, 125–132 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Evans, P. R. & Murshudov, G. N. How good are my data and what is the resolution? Acta Crystallogr. D 69, 1204–1214 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Agirre, J. et al. The CCP4 suite: integrative software for macromolecular crystallography. Acta Crystallogr. D 79, 449–461 (2023).

    ADS  CAS  Google Scholar 

  54. Tickle, I. J. et al. STARANISO (Global Phasing Ltd, 2018).

  55. Mirdita, M. et al. ColabFold: making protein folding accessible to all. Nat. Methods 19, 679–682 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  57. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Murshudov, G. N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D 67, 355–367 (2011).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    ADS  CAS  PubMed  Google Scholar 

  60. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    CAS  PubMed  Google Scholar 

  61. Agirre, J. et al. Privateer: software for the conformational validation of carbohydrate structures. Nat. Struct. Mol. Biol. 22, 833–834 (2015).

    CAS  PubMed  Google Scholar 

  62. Krissinel, E. & Henrick, K. Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 372, 774–797 (2007).

    CAS  PubMed  Google Scholar 

  63. Landau, M. et al. ConSurf 2005: the projection of evolutionary conservation scores of residues on protein structures. Nucleic Acids Res. 33, W299–W302 (2005).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gucwa, M. et al. CMM—an enhanced platform for interactive validation of metal binding sites. Protein Sci. 32, e4525 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Sonnenburg, E. D. et al. Specificity of polysaccharide use in intestinal bacteroides species determines diet-induced microbiota alterations. Cell 141, 1241–1252 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by operating grants from the Natural Sciences and Engineering Research Council of Canada (NSERC) and the Canadian Institutes of Health Research (CIHR) as well as infrastructure funds from the Canada Foundation for Innovation and BC Knowledge Development Fund. S.A.N. was supported by a Vanier scholarship from NSERC; A.C.L. by a GAP scholarship from the UBC Centre for Blood Research; and L.B. by an Early Postdoc Mobility grant from Swiss National Science Foundation. N.C.J.S. is a Tier I Canada Research Chair in Structure-guided Antibiotic Discovery. Part of the research described in this paper was performed using beamline CMCF-BM at the Canadian Light Source, a national research facility of the University of Saskatchewan, which is supported by the Canada Foundation for Innovation (CFI), the Natural Sciences and Engineering Research Council (NSERC), the National Research Council (NRC), the Canadian Institutes of Health Research (CIHR), the Government of Saskatchewan and the University of Saskatchewan. We thank the GM/CA beamline staff at beamline 23-ID-B at the APS for access and support. GM/CA@APS has been funded in whole or in part by Federal funds from the National Cancer Institute (ACB-12002) and the National Institute of General Medical Sciences (AGM-12006). We thank A. M. Deutschbauer for providing access to the B.T. mutant strains18; T. Warren for the initial inspiration for these studies; P. Rahfeld for guidance about the screening process; A. Noonan and S. Hallam for guidance with processing of the sequencing data; M. Ezhova for assistance with the NMR experiments; and F. Liu, S. Macdonald, P. Danby, B. Herring, C. Olagnon, J. Wardman, R. Bains and Y. Tian for suggestions and discussions.

Author information

Authors and Affiliations

Authors

Contributions

S.G.W., S.A.N. and L.B. conceived and developed the screen. Screening was performed by I.L.L. and S.A.N.; S.A.N., C.Y.Z. and E.B. performed the cloning, purification and biochemical characterization of the enzymes. S.A.N., I.L.L., C.Y.Z. and H.-M.C. synthesized all of the chemicals. A.C.L., L.S., L.J.W. and N.C.J.S. performed the crystallography analysis. S.A.N. and D.P. performed microbial growth and RT–qPCR assays under the guidance of H.B.; S.A.N., A.C.L. and S.G.W. wrote the manuscript with input from all of the authors.

Corresponding author

Correspondence to Stephen G. Withers.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Sebastian Hiller 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 figures and tables

Extended Data Fig. 1 Reactions catalysed by AL/BT3 and AL/BT1.

1H-NMR characterization of the reactions of a) AL3 and b) AL1.

Extended Data Fig. 2 Reaction catalysed by AL/BT2 and proposed transition states for AL/BT 1 and 2.

a) 1H-NMR characterization of the reaction of BT2 and b) proposed transition states for AL/BT1 (top) and AL/BT2 (bottom), B refers to protein residues capable of acid/base catalysis and M refers to the metal ions that coordinate the 3-keto groups.

Extended Data Fig. 3 Additional structural figures for AL/BT 1-3.

a) Active site of BT1 bound with substrate analogue 3k-1,5-anhydroglucitol with coordinating residues labelled. b) Active site of AL1 bound with glucose with coordinating residues labelled. c) Metal binding site of apo AL1 with coordinating residues labelled. d) Front view of overall fold of BT2 with jelly roll in orange, fingers (purple) labelled F1-F5, and active site denoted with a dashed circle. e) Metal binding site of apo BT2 with coordinating residues labelled. f) BT2 active site with conserved possible active site residues labelled. g) Active site of AL3 bound with trehalose with key residues labelled. h) Binding sites of trehalose observed in AL3 crystal soaked with trehalose. Active site is labelled as 1 and circled, secondary sites are labelled 2–6.

Extended Data Fig. 4 In vivo hydrolysis of substrates by Bacteroides species.

a) Homologues of the proteins from Bacteroides thetaiotaomicron in some other Bacteroides strains. The percent similarity of each of the encoded proteins to the corresponding BT homologue is shown in white. b) The percent of hydrolysed selective non-Koshland substrates 1 and 3 by different Bacteroides strains. All the strains were grown anaerobically and incubated with the substrates overnight. Bars represent the mean and error bars represent standard deviations (n = 3 technical replicates, derived from the same original culture). c) Transcriptional response of BT genes BT2156-60 to trehalose and sucrose as sole carbon sources. Representative genes from the fructan utilization locus (BT1759 and BT1763, encoding GH32 and SusC homologues, respectively) serve as positive controls for sucrose regulation65. Data were acquired at mid-log phase (9 h for trehalose, 5 h for sucrose) and are normalized to transcript levels prior to inoculating cells into the respective carbohydrate medium. Bars represent the mean and error bars represent the standard error of the mean (n = 4 replicates, two technical replicates each derived from two separate original cultures). d) Resulting fluorescence from hydrolysis of substrates 1 and 3 by B. thetaiotaomicron cells grown in different media. Growth on α,α-trehalose results in significantly higher activities while growth on glucose represses them, (n = 3 technical replicates, derived from the same original pre-culture, data for all replicates are shown.) e) Growth of B. thetaiotaomicron cells on cellobiose in absence and presence of trehalose, performed in triplicate for all the conditions shown here. f) hydrolysis of substrates 1 and 3 by B. thetaiotaomicron cells grown on different glycosides as the sole carbon source. Bars represent the mean and error bars represent standard deviations (n = 3 technical replicates, derived from the same original culture).

Source Data

Extended Data Fig. 5 Homologues of the proteins from P1C11 in bacteria from different environments.

Homologues of the proteins from P1C11 (Alistipes sp.) identified in bacteria from various environments (the source from which each bacterium is isolated is noted in parentheses below the name of bacterium). The percent similarity of each of the encoded proteins to the corresponding Alistipes homologue is shown in white. Dark blue denotes oxidoreductases that are not from the same family as AL3/AL4 and denoted in yellow are commonly co-occurring genes annotated as a DoxX-domain-containing protein, which is reported to form membrane-associated oxidoreductase complexes. Broken lines mean that a few genes are in between those shown and light grey shows other genes. The length of the arrows and the space between them does not correspond to the actual length of the DNA fragments in this figure. This is a representative list not an exhaustive one.

Extended Data Fig. 6 Homologues of the proteins from P2B11 in bacteria from different environments.

Homologues of the proteins from P2B11 identified in bacteria from various environments (the source from which each bacterium is isolated is noted in parentheses below the name of bacterium). The percent similarity of each of the encoded proteins to the corresponding P2B11 homologue is shown in white. Broken lines mean that a few genes are in between those shown and light grey shows other genes. The length of the arrows and the space between them does not correspond to the actual length of the DNA fragments in this figure. This is a representative list not an exhaustive one.

Extended Data Table 1 (Pseudo)-Michaelis-Menten kinetic parameters for enzymes
Extended Data Table 2 Data collection and refinement statistics

Supplementary information

Supplementary Information

Supplementary Methods, Figures, Tables and Schemes, as well as synthetic procedure and characterization for all compounds and nucleotide and amino acid sequences.

Reporting Summary

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

Nasseri, S.A., Lazarski, A.C., Lemmer, I.L. et al. An alternative broad-specificity pathway for glycan breakdown in bacteria. Nature 631, 199–206 (2024). https://doi.org/10.1038/s41586-024-07574-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07574-y

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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