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.

  • Letter
  • Published:

β-Barrel proteins tether the outer membrane in many Gram-negative bacteria

Abstract

Gram-negative bacteria have a cell envelope that comprises an outer membrane (OM), a peptidoglycan (PG) layer and an inner membrane (IM)1. The OM and PG are load-bearing, selectively permeable structures that are stabilized by cooperative interactions between IM and OM proteins2,3. In Escherichia coli, Braun’s lipoprotein (Lpp) forms the only covalent tether between the OM and PG and is crucial for cell envelope stability4; however, most other Gram-negative bacteria lack Lpp so it has been assumed that alternative mechanisms of OM stabilization are present5. We used a glycoproteomic analysis of PG to show that β-barrel OM proteins are covalently attached to PG in several Gram-negative species, including Coxiella burnetii, Agrobacterium tumefaciens and Legionella pneumophila. In C. burnetii, we found that four different types of covalent attachments occur between OM proteins and PG, with tethering of the β-barrel OM protein BbpA becoming most abundant in the stationary phase and tethering of the lipoprotein LimB similar throughout the cell cycle. Using a genetic approach, we demonstrate that the cell cycle-dependent tethering of BbpA is partly dependent on a developmentally regulated L,D-transpeptidase (Ldt). We use our findings to propose a model of Gram-negative cell envelope stabilization that includes cell cycle control and an expanded role for Ldts in covalently attaching surface proteins to PG.

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: β-barrel OM proteins are covalently attached to PG and form a tight tether in C. burnetii.
Fig. 2: β-barrel tethering in C. burnetii is abundant in stationary phase.
Fig. 3: The L,D-transpeptidase ldt2 is required for covalent attachment of BbpA and BbpB to PG.
Fig. 4: Model depicting attachments of OM proteins to PG in C. burnetii.

Similar content being viewed by others

Data availability

The MS datasets are available through the GlycoPOST repository (GPST000124). The cryo-EM micrographs used as data in Fig. 3c are available at Figshare (https://doi.org/10.6084/m9.figshare.12792506). The molecular dynamics simulation data and additional raw data that support the findings of this study are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

References

  1. Silhavy, T. J., Kahne, D. & Walker, S. The bacterial cell envelope. Cold Spring Harb. Perspect. Biol. 2, a000414 (2010).

    PubMed  PubMed Central  Google Scholar 

  2. Hwang, H., Paracini, N., Parks, J. M., Lakey, J. H. & Gumbart, J. C. Distribution of mechanical stress in the Escherichia coli cell envelope. Biochim. Biophys. Acta Biomembr. 1860, 2566–2575 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Rojas, E. R. et al. The outer membrane is an essential load-bearing element in Gram-negative bacteria. Nature 559, 617–621 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Asmar, A. T. & Collet, J.-F. Lpp, the Braun lipoprotein, turns 50—major achievements and remaining issues. FEMS Microbiol. Lett. 365, fny199 (2018).

    CAS  Google Scholar 

  5. Egan, A. J. F. Bacterial outer membrane constriction. Mol. Microbiol. 107, 676–687 (2018).

    CAS  PubMed  Google Scholar 

  6. Pagès, J. M., James, C. E. & Winterhalter, M. The porin and the permeating antibiotic: a selective diffusion barrier in Gram-negative bacteria. Nat. Rev. Microbiol. 6, 893–903 (2008).

    PubMed  Google Scholar 

  7. Galdiero, S. et al. Microbe–host interactions: structure and role of Gram-negative bacterial porins. Curr. Protein Pept. Sci. 13, 843–854 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Park, J. S. et al. Mechanism of anchoring of OmpA protein to the cell wall peptidoglycan of the gram-negative bacterial outer membrane. FASEB J. 26, 219–228 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Godlewska, R., Wiśniewska, K., Pietras, Z. & Jagusztyn-Krynicka, E. K. Peptidoglycan-associated lipoprotein (Pal) of Gram-negative bacteria: function, structure, role in pathogenesis and potential application in immunoprophylaxis. FEMS Microbiol. Lett. 298, 1–11 (2009).

    CAS  PubMed  Google Scholar 

  10. Samsudin, F., Ortiz-Suarez, M. L., Piggot, T. J., Bond, P. J. & Khalid, S. OmpA: a flexible clamp for bacterial cell wall attachment. Structure 24, 2227–2235 (2016).

    CAS  PubMed  Google Scholar 

  11. Braun, V. & Rehn, K. Chemical characterization, spatial distribution and function of a lipoprotein (murein-lipoprotein) of the E. coli cell wall. The specific effect of trypsin on the membrane structure. Eur. J. Biochem. 10, 426–438 (1969).

    CAS  PubMed  Google Scholar 

  12. Braun, V. Covalent lipoprotein from the outer membrane of Escherichia coli. Biochim. Biophys. Acta 415, 335–377 (1975).

    CAS  PubMed  Google Scholar 

  13. Magnet, S. et al. Identification of the L,D-transpeptidases responsible for attachment of the Braun lipoprotein to Escherichia coli peptidoglycan. J. Bacteriol. 189, 3927–3931 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Magnet, S., Dubost, L., Marie, A., Arthur, M. & Gutmann, L. Identification of the L,D-transpeptidases for peptidoglycan cross-linking in Escherichia coli. J. Bacteriol. 190, 4782–4785 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Cohen, E. J., Ferreira, J. L., Ladinsky, M. S., Beeby, M. & Hughes, K. T. Nanoscale-length control of the flagellar driveshaft requires hitting the tethered outer membrane. Science 356, 197–200 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Asmar, A. T. et al. Communication across the bacterial cell envelope depends on the size of the periplasm. PLoS Biol. 15, e2004303 (2017).

    PubMed  PubMed Central  Google Scholar 

  17. Butler, C. A. & Hoffman, P. S. Characterization of a major 31-kilodalton peptidoglycan-bound protein of Legionella pneumophila. J. Bacteriol. 172, 2401–2407 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Amano, K. & Williams, J. C. Partial characterization of peptidoglycan-associated proteins of Legionella pneumophila. J. Biochem. 94, 601–606 (1983).

    CAS  PubMed  Google Scholar 

  19. Amano, K. & Williams, J. C. Peptidoglycan of Legionella pneumophila: apparent resistance to lysozyme hydrolysis correlates with a high degree of peptide cross-linking. J. Bacteriol. 153, 520–526 (1983).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hoffman, P. S., Seyer, J. H. & Butler, C. A. Molecular characterization of the 28- and 31-kilodalton subunits of the Legionella pneumophila major outer membrane protein. J. Bacteriol. 174, 908–913 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Amano, K. & Williams, J. C. Sensitivity of Coxiella burnetii peptidoglycan to lysozyme hydrolysis and correlation of sacculus rigidity with peptidoglycan-associated proteins. J. Bacteriol. 160, 989–993 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Barton, I. S., Fuqua, C. & Platt, T. G. Ecological and evolutionary dynamics of a model facultative pathogen: Agrobacterium and crown gall disease of plants. Environ. Microbiol. 20, 16–29 (2018).

    PubMed  Google Scholar 

  23. Oliva, G., Sahr, T. & Buchrieser, C. The life cycle of L. pneumophila: cellular differentiation is linked to virulence and metabolism. Front. Cell. Infect. Microbiol. 8, 3 (2018).

    PubMed  PubMed Central  Google Scholar 

  24. Minnick, M. F. & Raghavan, R. Developmental biology of Coxiella burnetii. Adv. Exp. Med. Biol. 984, 231–248 (2012).

    CAS  PubMed  Google Scholar 

  25. Bern, M., Beniston, R. & Mesnage, S. Towards an automated analysis of bacterial peptidoglycan structure. Anal. Bioanal. Chem. 409, 551–560 (2017).

    CAS  PubMed  Google Scholar 

  26. Braun, V. & Wolff, H. The murein-lipoprotein linkage in the cell wall of Escherichia coli. Eur. J. Biochem. 14, 387–391 (1970).

    CAS  PubMed  Google Scholar 

  27. Battisti, J. M., Hicks, L. D. & Minnick, M. F. A unique Coxiella burnetii lipoprotein involved in metal binding (LimB). Microbiology (Reading) 157, 966–976 (2011).

    CAS  Google Scholar 

  28. Braun, V. & Bosch, V. Sequence of the murein-lipoprotein and the attachment site of the lipid. Eur. J. Biochem. 28, 51–69 (1972).

    CAS  PubMed  Google Scholar 

  29. Pavlova, A., Hwang, H., Lundquist, K., Balusek, C. & Gumbart, J. C. Living on the edge: simulations of bacterial outer-membrane proteins. Biochim. Biophys. Acta 1858, 1753–1759 (2016).

    CAS  PubMed  Google Scholar 

  30. McCaul, T. F. & Williams, J. C. Developmental cycle of Coxiella burnetii: structure and morphogenesis of vegetative and sporogenic differentiations. J. Bacteriol. 147, 1063–1076 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Sandoz, K. M. et al. Transcriptional profiling of Coxiella burnetii reveals extensive cell wall remodeling in the small cell variant developmental form. PLoS ONE 11, e0149957 (2016).

    PubMed  PubMed Central  Google Scholar 

  32. Cameron, T. A., Anderson-Furgeson, J., Zupan, J. R., Zik, J. J. & Zambryski, P. C. Peptidoglycan synthesis machinery in Agrobacterium tumefaciens during unipolar growth and cell division. mBio 5, e01219-14 (2014).

    PubMed  PubMed Central  Google Scholar 

  33. Morè, N. et al. Peptidoglycan remodeling enables Escherichia coli to survive severe outer membrane assembly defect. mBio 10, e02729-18 (2019).

    PubMed  PubMed Central  Google Scholar 

  34. Sonntag, I., Schwarz, H., Hirota, Y. & Henning, U. Cell envelope and shape of Escherichia coli: multiple mutants missing the outer membrane lipoprotein and other major outer membrane proteins. J. Bacteriol. 136, 280–285 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Bertrand, R. L. Lag phase is a dynamic, organized, adaptive, and evolvable period that prepares bacteria for cell division. J. Bacteriol. 201, e00697-18 (2019).

    PubMed  PubMed Central  Google Scholar 

  36. Delcour, A. H. Outer membrane permeability and antibiotic resistance. Biochim. Biophys. Acta 1794, 808–816 (2009).

    CAS  PubMed  Google Scholar 

  37. Mainardi, J.-L. et al. A novel peptidoglycan cross-linking enzyme for a β-lactam-resistant transpeptidation pathway. J. Biol. Chem. 280, 38146–38152 (2005).

    CAS  PubMed  Google Scholar 

  38. Sutterlin, L., Edoo, Z., Hugonnet, J.-E., Mainardi, J.-L. & Arthur, M. Peptidoglycan cross-linking activity of L,D-transpeptidases from Clostridium difficile and inactivation of theses enzymes by β-lactams. Antimicrob. Agents Chemother. 62, e01607-17 (2017).

    PubMed  PubMed Central  Google Scholar 

  39. Zhao, H., Patel, V., Helmann, J. D. & Dorr, T. Don’t let sleeping dogmas lie: new views of peptidoglycan synthesis and its regulation. Mol. Microbiol. 106, 847–860 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Bergkessel, M., Basta, D. W. & Newman, D. K. The physiology of growth arrest: uniting molecular and environmental microbiology. Nat. Rev. Microbiol. 14, 549–562 (2016).

    CAS  PubMed  Google Scholar 

  41. Beare, P. A., Larson, C. L., Gilk, S. D. & Heinzen, R. A. Two systems for targeted gene deletion in Coxiella burnetii. Appl. Environ. Microbiol. 78, 4580–4589 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Sandoz, K. M., Beare, P. A., Cockrell, D. C. & Heinzen, R. A. Complementation of arginine auxotrophy for genetic transformation of Coxiella burnetii by use of a defined axenic medium. Appl. Environ. Microbiol. 82, 3042–3051 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Cangelosi, G. A., Best, E. A., Martinetti, G. & Nester, E. W. Genetic analysis of Agrobacterium. Methods Enzymol. 204, 384–397 (1991).

    CAS  PubMed  Google Scholar 

  44. Söding, J. Protein homology detection by HMM-HMM comparison. Bioinformatics 21, 951–960 (2005).

    PubMed  Google Scholar 

  45. Zimmermann, L. et al. A completely reimplemented MPI bioinformatics toolkit with a new HHpred server at its core. J. Mol. Biol. 430, 2237–2243 (2018).

    CAS  PubMed  Google Scholar 

  46. Pautsch, A. & Schulz, G. E. High-resolution structure of the OmpA membrane domain. J. Mol. Biol. 298, 273–282 (2000).

    CAS  PubMed  Google Scholar 

  47. Kamisetty, H., Ovchinnikov, S. & Baker, D. Assessing the utility of coevolution-based residue–residue contact predictions in a sequence- and structure-rich era. Proc. Natl Acad. Sci. USA 110, 15674–15679 (2013).

    CAS  PubMed  Google Scholar 

  48. Balakrishnan, S., Kamisetty, H., Carbonell, J. G., Lee, S.-I. & Langmead, C. J. Learning generative models for protein fold families. Proteins 79, 1061–1078 (2011).

    CAS  PubMed  Google Scholar 

  49. Remmert, M., Biegert, A., Hauser, A. & Söding, J. HHblits: lightning-fast iterative protein sequence searching by HMM-HMM alignment. Nat. Methods 9, 173–175 (2011).

    PubMed  Google Scholar 

  50. UniProt Consortium. UniProt: a worldwide hub of protein knowledge. Nucleic Acids Res. 47, D506–D515 (2019).

  51. Ovchinnikov, S. et al. Protein structure determination using metagenome sequence data. Science 355, 294–298 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Song, Y. et al. High-resolution comparative modeling with RosettaCM. Structure 21, 1735–1742 (2013).

    CAS  PubMed  Google Scholar 

  53. Viklund, H. & Elofsson, A. OCTOPUS: improving topology prediction by two-track ANN-based preference scores and an extended topological grammar. Bioinformatics 24, 1662–1668 (2008).

    CAS  Google Scholar 

  54. Vandeputte-Rutten, L., Bos, M. P., Tommassen, J. & Gros, P. Crystal structure of Neisserial surface protein A (NspA), a conserved outer membrane protein with vaccine potential. J. Biol. Chem. 278, 24825–24830 (2003).

    CAS  PubMed  Google Scholar 

  55. Zamboni, D. S. et al. Stimulation of toll-like receptor 2 by Coxiella burnetii is required for macrophage production of pro-inflammatory cytokines and resistance to infection. J. Biol. Chem. 279, 54405–54415 (2004).

    CAS  PubMed  Google Scholar 

  56. Narasaki, C. T. & Toman, R. Lipopolysaccharide of Coxiella burnetii. Adv. Exp. Med. Biol. 984, 65–90 (2012).

    CAS  PubMed  Google Scholar 

  57. Stead, C. M., Cockrell, D. C., Beare, P. A., Miller, H. E. & Heinzen, R. A. A Coxiella burnetii phospholipase A homolog pldA is required for optimal growth in macrophages and developmental form lipid remodeling. BMC Microbiol. 18, 33 (2018).

    PubMed  PubMed Central  Google Scholar 

  58. Gumbart, J. C., Beeby, M., Jensen, G. J. & Roux, B. Escherichia coli peptidoglycan structure and mechanics as predicted by atomic-scale simulations. PLoS Comput. Biol. 10, e1003475 (2014).

    PubMed  PubMed Central  Google Scholar 

  59. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996).

    CAS  PubMed  Google Scholar 

  60. Phillips, J. C. et al. Scalable molecular dynamics with NAMD. J. Comput. Chem. 26, 1781–1802 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Darden, T., York, D. & Pedersen, L. Particle mesh Ewald: an N log(N) method for Ewald sums in large systems. J. Chem. Phys. 98, 10089–10092 (1993).

    CAS  Google Scholar 

  62. Huang, J. et al. CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14, 71–73 (2017).

    CAS  PubMed  Google Scholar 

  63. Klauda, J. B. et al. Update of the CHARMM all-atom additive force field for lipids: validation on six lipid types. J. Phys. Chem. B 114, 7830–7843 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Suzuki, A. E. Acosta Martin and M. Collins for their contribution to developing the MS method and V. Nair for his help with cryo-EM. We thank R. Kissinger for his three-dimensional modelling, with assistance from A. Athman, and A. Mora for graphics support. We thank X. De Bolle and P. Godessart for stimulating discussion and J. Zupan, P. Zambryski, D. Kelly, A. Taylor, J. Shaw, E. F. Diaz Parga, R. Wheeler, I. Boneca, M.-K. Taha and E. Hoiczyk for the bacterial cultures used for peptidoglycan extraction. The PG MS analyses were performed by the biOMICS Facility of the Faculty of Science Mass Spectrometry Centre at the University of Sheffield, UK and the Research Technologies Branch at the National Institutes of Health (NIH) in Rockville, USA. The cryo-EM work was performed at the Research Technologies Branch at the NIH in Hamilton, USA. This work was supported by the Intramural Research Program of the NIH, National Institute of Allergy and Infectious Diseases (R.A.H. and S.A.P.), a Medical Research Council grant no. MR/S009272/1 (S.M.) and a Biotechnology and Biological Sciences Research Council grant no. BBSRC BB/N000951/1 (S.M.). R.E.S. and A.V.P. are supported by a Biotechnology and Biological Sciences Research Council studentship (doctoral training program grant no. BB/M011151/1). J.C.G. acknowledges support from the NIH under grant no. R01-GM123169. C.J.C. was supported by a National Science Foundation (NSF) Graduate Research Fellowship under grant no. 2017219379. J.M.P. was supported by the Laboratory Directed Research and Development program at Oak Ridge National Laboratory, which is managed by UT-Battelle for the US Department of Energy under contract no. DE-AC05-00OR22725. This work used resources of the Compute and Data Environment for Science at Oak Ridge National Laboratory as well as the Extreme Science and Engineering Discovery Environment (allocation no. TG-MCB130173), which is supported by an NSF grant no. ACI-1548562.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed instrumentally. K.M.S., R.E.S., A.V.P., M.B., R.A.M. and S.M. designed and performed the experiments and MS/MS data analysis. P.A.B. generated the recombinant and knockout strains. J.M.P., C.J.C., J.C.G. and H.H. performed the protein modelling and molecular dynamics simulation. S.M., S.A.P. and R.A.H. supervised the study. K.M.S., S.M. and R.A.H. prepared the manuscript.

Corresponding authors

Correspondence to Kelsi M. Sandoz, Stéphane Mesnage or Robert A. Heinzen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 PG modifications identified on proteobacterial OM proteins.

Proteins from A. tumefaciens, C. burnetii, and L. pneumophila are covalently attached to PG. Proteins identified in MS/MS data that contained a PG modification scoring higher than the first decoy and containing greater than 40% of the expected b- and y- ions were established as cut-off criteria. Predicted signal peptide cleavage sites of OM proteins covalently attached to PG from C. burnetii, L. pneumophila, and A. tumefaciens are shown. Amino acid numbering is based on the predicted signal peptide cleavage site. Peptide sequences that were found covalently attached to PG are bolded. Residues with a PG tripeptide modification are also highlighted in red.

Extended Data Fig. 2 Automated spectrum assignment identifies PG modifications on C. burnetii OM proteins.

Representative MS/MS spectra for C. burnetii BbpA, ala-BbpA, BbpB β-barrel proteins and the lipoprotein LimB, covalently attached to mDAP (m) residues of PG. Spectra are shown as annotated by Byonic with manual annotations corresponding to internal fragments in black. J[ + 72.0848] has been replaced by m to represent mDAP. The PG tripeptide (AEm) is highlighted in red in the HCD spectra showing covalent attachment of PG to BbpA, ala-BbpA, and BbpB. The LimB Lys21 residue with PG tripeptide modification is highlighted in red in the ETD spectra showing covalent attachment of PG to LimB.

Extended Data Fig. 3 Automated spectrum assignment identifies PG modifications on β-barrel proteins from A. tumefaciens.

Representative MS/MS spectra for A. tumefaciens β-barrel proteins covalently attached to mDAP (m) residues of PG. Residues with PG tripeptide modification are highlighted in red. Spectra are shown as annotated by Byonic using an unbiased search approach.

Extended Data Fig. 4 Automated spectrum assignment identifies PG modifications on β-barrel proteins from L. pneumophila.

Representative MS/MS spectra for L. pneumophila Major Outer Membrane Protein (MOMP) covalently attached to mDAP (m) residues of PG. Residues with PG tripeptide modification are highlighted in red. Spectra are shown as annotated by Byonic using an unbiased search approach.

Extended Data Fig. 5 Structural topology of C. burnetii BbpA and BbpB.

Predicted periplasmic, transmembrane, and extracellular domains of BbpA and BbpB using PRED-TMBB. BbpA and BbpB are depicted with periplasmic N-terminal regions. Similar topology is predicted in β-barrel proteins from A. tumefaciens and L. pneumophila that are covalently attached to PG.

Extended Data Fig. 6 Structural modeling of C. burnetii OM proteins.

a, The protein sequences of BbpA, BbpB, and LimB are shown without N-terminal signal peptides. The disordered N-terminal domains of BbpA and BbpB were excluded from structural models but were included in subsequent molecular dynamics simulations (blue). Residues bound to PG in molecular dynamics simulations are highlighted in red. b, The mature LimB was modeled as a random coil lacking any appreciable secondary structure. The three acyl tails at the N-terminus are shown in grey as is Lys21. c, Contact map predicted from coevolution analysis of BbpA. Contacts are shown in shades of blue (darker blue = higher probability) and contacts from E. coli OmpA (PDB entry 1QJP) are shown in gray. d, Predicted contacts mapped onto the final Rosetta model of BbpA. Contacts are color coded by Cα-Cα distance: green (<5 Å), yellow (5-10 Å), red (>10 Å).

Extended Data Fig. 7 β-barrels form a tight tether between the OM and PG.

Structural model of BbpA (red) in the C. burnetii OM. Structures of the cell envelope are colored as follows: inner leaflet of OM (grey), lipid A of LPS (orange), core oligosaccharides (yellow), glycan chains of PG (blue) peptide stems of PG (green). A similar model was generated for BbpB. b, Molecular dynamics simulation of C. burnetii OM-PG protein-tethered models. The distance in angstroms between the phosphorus atoms of the inner leaflet of the OM and PG layers was measured for three runs for BbpA, ala-BbpA, BbpB, LimB, and Lpp from E. coli. The solid lines are running averages. Distances measured in angstroms for run 1, run 2, and run 3 are shown.

Extended Data Fig. 8 Predicted or annotated Ldts in Proteobacteria used in this study.

Predicted or annotated Ldts in E. coli (lpp + ), C. burnetii, L. pneumophila, A. tumefaciens, M. xanthus, N. gonorrhoeae, H. pylori, and C. jejuni. The latter seven organisms lack lpp homologs. The C. burnetii ldt genes that were successfully inactivated in this study are denoted with an asterisk. C. burnetii ldts upregulated during stationary phase are in bold. The following ldts were previously annotated as enhanced entry genes (enh); cbu0053, cbu0318, cbu1122, cbu1138, and cbu1394.

Extended Data Fig. 9 The L,D transpeptidase ldt2 is required for covalent attachment of BbpA and BbpB to PG.

XIC analysis was performed on PG that was extracted from WT and ∆ldt mutant strains and analyzed by MS/MS. XIC’s of precursor masses (m/z) corresponding to PG-bound BbpA (AEmGGPDYVPAPS, m/z = 906.909, z = 2), ala-BbpA (AEmAGGPDYVPAPS, m/z = 942.427, z = 2), and BbpB (AEmGGPDIPM, m/z = 769.843, z = 2) are shown.

Extended Data Fig. 10 Onset of replication is delayed in the ∆ldt2 deletion mutant.

Growth of wild-type (WT) C. burnetii and a ∆ldt2 mutant strain in ACCM-D was assessed using qPCR to quantitate genome equivalents (GE) during a 21-day incubation. The results are expressed as the means from n = 3 independent experiments. Error bars indicate the standard error of the mean, and asterisks indicate a statistically significant difference from WT C. burnetii. **P at 3, 6 and 9 days post-inoculation = 0.0046, 0.0027, and 0.0001, respectively. Statistical significance was calculated using a two-sided Student’s t test.

Supplementary information

Source data

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sandoz, K.M., Moore, R.A., Beare, P.A. et al. β-Barrel proteins tether the outer membrane in many Gram-negative bacteria. Nat Microbiol 6, 19–26 (2021). https://doi.org/10.1038/s41564-020-00798-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-00798-4

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