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:

Staphylococcus aureus cell growth and division are regulated by an amidase that trims peptides from uncrosslinked peptidoglycan

Abstract

Bacteria are protected by a polymer of peptidoglycan that serves as an exoskeleton1. In Staphylococcus aureus, the peptidoglycan assembly enzymes relocate during the cell cycle from the periphery, where they are active during growth, to the division site where they build the partition between daughter cells2,3,4. But how peptidoglycan synthesis is regulated throughout the cell cycle is poorly understood5,6. Here, we used a transposon screen to identify a membrane protein complex that spatially regulates S. aureus peptidoglycan synthesis. This complex consists of an amidase that removes stem peptides from uncrosslinked peptidoglycan and a partner protein that controls its activity. Amidases typically hydrolyse crosslinked peptidoglycan between daughter cells so that they can separate7. However, this amidase controls cell growth. In its absence, peptidoglycan synthesis becomes spatially dysregulated, which causes cells to grow so large that cell division is defective. We show that the cell growth and division defects due to loss of this amidase can be mitigated by attenuating the polymerase activity of the major S. aureus peptidoglycan synthase. Our findings lead to a model wherein the amidase complex regulates the density of peptidoglycan assembly sites to control peptidoglycan synthase activity at a given subcellular location. Removal of stem peptides from peptidoglycan at the cell periphery promotes peptidoglycan synthase relocation to midcell during cell division. This mechanism ensures that cell expansion is properly coordinated with cell division.

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: LytH is an amidase involved in cell growth and division.
Fig. 2: The LytH–ActH complex cleaves stem peptides from uncrosslinked peptidoglycan.
Fig. 3: Slowing peptidoglycan synthesis compensates for loss of LytH.
Fig. 4: LytH spatially regulates peptidoglycan synthesis.

Similar content being viewed by others

Data availability

Transposon sequencing data (accession numbers SAMN08025141 and SAMN08025168) can be found in the NCBI BioSample database. Whole-genome sequencing data (accession number PRJNA579395) can be found in the NCBI BioProject database. All other data are available in the manuscript, Extended Data, Supplementary Information or Source Data.

Code availability

Code is available at https://github.com/SuzanneWalkerLab/imageanalysis.

References

  1. Vollmer, W., Blanot, D. & de Pedro, M. A. Peptidoglycan structure and architecture. FEMS Microbiol. Rev. 32, 149–167 (2008).

    CAS  PubMed  Google Scholar 

  2. Pinho, M. G., Kjos, M. & Veening, J.-W. How to get (a)round: mechanisms controlling growth and division of coccoid bacteria. Nat. Rev. Microbiol. 11, 601–614 (2013).

    CAS  PubMed  Google Scholar 

  3. Monteiro, J. M. et al. Peptidoglycan synthesis drives an FtsZ-treadmilling-independent step of cytokinesis. Nature 554, 528–532 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Lund, V. A. et al. Molecular coordination of Staphylococcus aureus cell division. eLife 7, e32057 (2018).

    PubMed  PubMed Central  Google Scholar 

  5. Typas, A., Banzhaf, M., Gross, C. A. & Vollmer, W. From the regulation of peptidoglycan synthesis to bacterial growth and morphology. Nat. Rev. Microbiol. 10, 123–136 (2011).

    PubMed  PubMed Central  Google Scholar 

  6. Egan, A. J. F., Cleverley, R. M., Peters, K., Lewis, R. J. & Vollmer, W. Regulation of bacterial cell wall growth. FEBS J. 284, 851–867 (2017).

    CAS  PubMed  Google Scholar 

  7. Vollmer, W., Joris, B., Charlier, P. & Foster, S. Bacterial peptidoglycan (murein) hydrolases. FEMS Microbiol. Rev. 32, 259–286 (2008).

    CAS  PubMed  Google Scholar 

  8. Sauvage, E., Kerff, F., Terrak, M., Ayala, J. A. & Charlier, P. The penicillin-binding proteins: structure and role in peptidoglycan biosynthesis. FEMS Microbiol. Rev. 32, 234–258 (2008).

    CAS  PubMed  Google Scholar 

  9. Adams, D. W. & Errington, J. Bacterial cell division: assembly, maintenance and disassembly of the Z ring. Nat. Rev. Microbiol. 7, 642–653 (2009).

    CAS  PubMed  Google Scholar 

  10. Veiga, H., Jorge, A. M. & Pinho, M. G. Absence of nucleoid occlusion effector Noc impairs formation of orthogonal FtsZ rings during Staphylococcus aureus cell division. Mol. Microbiol. 80, 1366–1380 (2011).

    CAS  PubMed  Google Scholar 

  11. Jorge, A. M., Hoiczyk, E., Gomes, J. P. & Pinho, M. G. EzrA contributes to the regulation of cell size in Staphylococcus aureus. PLoS ONE 6, e27542 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Pang, T., Wang, X., Lim, H. C., Bernhardt, T. G. & Rudner, D. Z. The nucleoid occlusion factor Noc controls DNA replication initiation in Staphylococcus aureus. PLoS Genet. 13, e1006908 (2017).

    PubMed  PubMed Central  Google Scholar 

  13. Santiago, M. et al. Genome-wide mutant profiling predicts the mechanism of a lipid II binding antibiotic. Nat. Chem. Biol. 14, 601–608 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Tomasz, A., Albino, A. & Zanati, E. Multiple antibiotic resistance in a bacterium with suppressed autolytic system. Nature 227, 138–140 (1970).

    CAS  PubMed  Google Scholar 

  15. Oshida, T. et al. A Staphylococcus aureus autolysin that has an N-acetylmuramoyl-l-alanine amidase domain and an endo-beta-N-acetylglucosaminidase domain: cloning, sequence analysis, and characterization. Proc. Natl Acad. Sci. USA 92, 285–289 (1995).

    CAS  PubMed  Google Scholar 

  16. Kajimura, J. et al. Identification and molecular characterization of an N-acetylmuramyl-l-alanine amidase Sle1 involved in cell separation of Staphylococcus aureus. Mol. Microbiol. 58, 1087–1101 (2005).

    CAS  PubMed  Google Scholar 

  17. Lenz, J. D. et al. Amidase activity of AmiC controls cell separation and stem peptide release and is enhanced by NlpD in Neisseria gonorrhoeae. J. Biol. Chem. 291, 10916–10933 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Rocaboy, M. et al. The crystal structure of the cell division amidase AmiC reveals the fold of the AMIN domain, a new peptidoglycan binding domain. Mol. Microbiol. 90, 267–277 (2013).

    CAS  PubMed  Google Scholar 

  19. Kawata, S., Takemura, T. & Yokogawa, K. Characterization of two N-acetylmuramidases from Streptomyces globisporus 1829. Agr. Biol. Chem. 47, 1501–1508 (1983).

    CAS  Google Scholar 

  20. Qiao, Y. et al. Lipid II overproduction allows direct assay of transpeptidase inhibition by β-lactams. Nat. Chem. Biol. 13, 793–798 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Rebets, Y. et al. Moenomycin resistance mutations in Staphylococcus aureus reduce peptidoglycan chain length and cause aberrant cell division. ACS Chem. Biol. 9, 459–467 (2014).

    CAS  PubMed  Google Scholar 

  22. Welsh, M. A. et al. Identification of a functionally unique family of penicillin-binding proteins. J. Am. Chem. Soc. 139, 17727–17730 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Schaefer, K., Matano, L. M., Qiao, Y., Kahne, D. & Walker, S. In vitro reconstitution demonstrates the cell wall ligase activity of LCP proteins. Nat. Chem. Biol. 13, 396–401 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lupoli, T. J. et al. Studying a cell division amidase using defined peptidoglycan substrates. J. Am. Chem. Soc. 131, 18230–18231 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Schaefer, K., Owens, T. W., Kahne, D. & Walker, S. Substrate preferences establish the order of cell wall assembly in Staphylococcus aureus. J. Am. Chem. Soc. 140, 2442–2445 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Lovering, A. L., de Castro, L. H., Lim, D. & Strynadka, N. C. J. Structural insight into the transglycosylation step of bacterial cell-wall biosynthesis. Science 315, 1402–1405 (2007).

    CAS  PubMed  Google Scholar 

  27. Kuru, E. et al. In situ probing of newly synthesized peptidoglycan in live bacteria with fluorescent d-amino acids. Angew. Chem. Int. Ed. Engl. 51, 12519–12523 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Pinho, M. G. & Errington, J. Dispersed mode of Staphylococcus aureus cell wall synthesis in the absence of the division machinery. Mol. Microbiol. 50, 871–881 (2003).

    CAS  PubMed  Google Scholar 

  29. Tan, C. M. et al. Restoring methicillin-resistant Staphylococcus aureus susceptibility to β-lactam antibiotics. Sci. Transl. Med. 4, 126ra35 (2012).

    PubMed  Google Scholar 

  30. Ruiz, N. Bioinformatics identification of MurJ (MviN) as the peptidoglycan lipid II flippase in Escherichia coli. Proc. Natl Acad. Sci. USA 105, 15553–15557 (2008).

    CAS  PubMed  Google Scholar 

  31. Sham, L.-T. et al. MurJ is the flippase of lipid-linked precursors for peptidoglycan biogenesis. Science 345, 220–222 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Pinho, M. G. & Errington, J. Recruitment of penicillin-binding protein PBP2 to the division site of Staphylococcus aureus is dependent on its transpeptidation substrates. Mol. Microbiol. 55, 799–807 (2005).

    CAS  PubMed  Google Scholar 

  33. Uehara, T., Parzych, K. R., Dinh, T. & Bernhardt, T. G. Daughter cell separation is controlled by cytokinetic ring-activated cell wall hydrolysis. EMBO J. 29, 1412–1422 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Sham, L.-T., Barendt, S. M., Kopecky, K. E. & Winkler, M. E. Essential PcsB putative peptidoglycan hydrolase interacts with the essential FtsXSpn cell division protein in Streptococcus pneumoniae D39. Proc. Natl Acad. Sci. USA 108, E1061–E1069 (2011).

    PubMed  Google Scholar 

  35. Bartual, S. G. et al. Structural basis of PcsB-mediated cell separation in Streptococcus pneumoniae. Nat. Commun. 5, 3842 (2014).

    CAS  PubMed  Google Scholar 

  36. Rued, B. E. et al. Structure of the large extracellular loop of FtsX and its interaction with the essential peptidoglycan hydrolase PcsB in Streptococcus pneumoniae. MBio 10, e02622-18 (2019).

    PubMed  PubMed Central  Google Scholar 

  37. Meisner, J. et al. FtsEX is required for CwlO peptidoglycan hydrolase activity during cell wall elongation in Bacillus subtilis. Mol. Microbiol. 89, 1069–1083 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Domínguez-Cuevas, P., Porcelli, I., Daniel, R. A. & Errington, J. Differentiated roles for MreB-actin isologues and autolytic enzymes in Bacillus subtilis morphogenesis. Mol. Microbiol. 89, 1084–1098 (2013).

    PubMed  PubMed Central  Google Scholar 

  39. Rather, P. Role of rhomboid proteases in bacteria. Biochim. Biophys. Acta 1828, 2849–2854 (2013).

    CAS  PubMed  Google Scholar 

  40. Maegawa, S., Ito, K. & Akiyama, Y. Proteolytic action of GlpG, a Rhomboid protease in the Escherichia coli cytoplasmic membrane. Biochemistry 44, 13543–13552 (2005).

    CAS  PubMed  Google Scholar 

  41. Liew, A. T. F. et al. A simple plasmid-based system that allows rapid generation of tightly controlled gene expression in Staphylococcus aureus. Microbiology 157, 666–676 (2011).

    CAS  PubMed  Google Scholar 

  42. Kato, F. & Sugai, M. A simple method of markerless gene deletion in Staphylococcus aureus. J. Microbiol. Methods 87, 76–81 (2011).

    CAS  PubMed  Google Scholar 

  43. Lee, W. et al. Antibiotic combinations that enable one-step, targeted mutagenesis of chromosomal genes. ACS Infect. Dis. 4, 1007–1018 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Charpentier, E. et al. Novel cassette-based shuttle vector system for Gram-positive bacteria. Appl. Environ. Microbiol. 70, 6076–6085 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. de Jong, N. W. M., van der Horst, T., van Strijp, J. A. G. & Nijland, R. Fluorescent reporters for markerless genomic integration in Staphylococcus aureus. Sci. Rep. 7, 43889 (2017).

    PubMed  PubMed Central  Google Scholar 

  46. Christie, G. E. et al. The complete genomes of Staphylococcus aureus bacteriophages 80 and 80α—implications for the specificity of SaPI mobilization. Virology 407, 381–390 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Qiao, Y. et al. Detection of lipid-linked peptidoglycan precursors by exploiting an unexpected transpeptidase reaction. J. Am. Chem. Soc. 136, 14678–14681 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Men, H., Park, P., Ge, M. & Walker, S. Substrate synthesis and activity assay for MurG. J. Am. Chem. Soc. 120, 2484–2485 (1998).

    CAS  Google Scholar 

  49. Tsukamoto, H. & Kahne, D. N-methylimidazolium chloride-catalyzed pyrophosphate formation: application to the synthesis of lipid I and NDP-sugar donors. Bioorg. Med. Chem. Lett. 21, 5050–5053 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Lee, W. et al. The mechanism of action of lysobactin. J. Am. Chem. Soc. 138, 100–103 (2016).

    CAS  PubMed  Google Scholar 

  51. Santiago, M. et al. A new platform for ultra-high density Staphylococcus aureus transposon libraries. BMC Genomics 16, 252 (2015).

    PubMed  PubMed Central  Google Scholar 

  52. Kühner, D., Stahl, M., Demircioglu, D. D. & Bertsche, U. From cells to muropeptide structures in 24 h: peptidoglycan mapping by UPLC–MS. Sci. Rep. 4, 7494 (2014).

    PubMed  PubMed Central  Google Scholar 

  53. Barrett, D. et al. Analysis of glycan polymers produced by peptidoglycan glycosyltransferases. J. Biol. Chem. 282, 31964–31971 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Monteiro, J. M. et al. Cell shape dynamics during the staphylococcal cell cycle. Nat. Commun. 6, 8055 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Meyer, F. Topographic distance and watershed lines. Signal Process. 38, 113–125 (1994).

    Google Scholar 

  56. Maurer, C. R., Qi, R. & Raghavan, V. A linear time algorithm for computing exact Euclidean distance transforms of binary images in arbitrary dimensions. IEEE Trans. Pattern Anal. Mach. Intell. 25, 265–270 (2003).

    Google Scholar 

  57. Rosenfeld, A. & Pfaltz, J. L. Sequential operations in digital picture processing. JACM 13, 471–494 (1966).

    Google Scholar 

  58. Paglieroni, D. W. Distance transforms: properties and machine vision applications. CVGIP Graph. Models Image Process. 54, 56–74 (1992).

    Google Scholar 

Download references

Acknowledgements

The authors thank M. Welsh and A. Taguchi for help with LC–MS analysis and protein expression. They thank T. Pang from the Bernhardt and Rudner laboratories for strain TD215. Fluorescence images were partially acquired at the MicRoN core at Harvard Medical School. The authors acknowledge support from the National Science Foundation (grant no. DGE1144152 to T.D.), the National Institutes of Health (grant nos. P01AI083214, R01GM076710, R01AI139011, R01AI099144), and the European Research Council (grant no. ERC-2017-CoG-771709 to M.G.P).

Author information

Authors and Affiliations

Authors

Contributions

S.W. and T.D. conceived the project, designed experiments and analysed the data with advice from M.G.P on acquisition and analysis of fluorescence images. T.D. performed all experiments except the radiolabelled gel assays, which were performed by K.S., and some of the microscopy experiments, which were performed by P.B.F. A.G.S. and T.D. developed scripts for image analysis. K.C. analysed the transposon sequencing data. S.W., D.K. and T.D. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Suzanne Walker.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Deletion of lytH sensitizes cells to oxacillin and causes growth defects.

a, HG003 strains were spotted on TSA containing 0.1 μg/mL oxacillin and 0.4 μM of the inducer, anhydrotetracycline (atc), when indicated. Strains expressing lytH or gfp from an atc- inducible promoter in a ∆lytH background are denoted by plytH or pgfp, respectively. b, Transmission electron micrographs: Additional examples of defects in placement of nascent septa observed for the ∆lytH mutant. Scale bars, 500 nm. c, Growth was assessed by measuring optical density (left graph) and colony-forming units (right graph). Data represent mean ± SD. Error bars that are smaller than the dots are not shown. Data are representative of two (a-b) and three (c) independent experiments.

Extended Data Fig. 2 Muropeptide analysis identifies a tetrasaccharide- monopeptide.

a, Schematic of sacculi isolation and mutanolysin/NaBH4 treatment to generate muropeptides for LC-MS analysis. b, Total ion chromatograms comparing the muropeptide species isolated from WT vs. ∆lytH sacculi. Muropeptide species eluted between 12-18 min using this analytical method. The XCMS online platform was used to identify species that were enriched or depleted between the two strains. c, The tetrasaccharide-monopeptide species was enriched in sacculi isolated from cells expressing lytHWT. A chemical structure consistent with this species, based on an [M + 2] ion with experimental m/z = 866.3881, is shown. d, The [M + 2] ion was targeted for fragmentation to confirm the species was a tetrasaccharide-monopeptide. e, The theoretical and experimental isotope distributions for the tetrasaccharide- monopeptide species are shown.

Extended Data Fig. 3 LytH pulled down ActH, a previously uncharacterized polytopic membrane protein.

a, A homology model of LytH highlights the predicted catalytic residues identified by sequence alignment of S. aureus LytH with E. coli AmiC. b, TMHMM topology prediction suggests that LytH is anchored in the cytoplasmic membrane. c, Schematic representation of the BDL-labeling assay to assess LytH hydrolytic activity in vitro. Short glycan strands were synthesized from Lipid II using S. aureus SgtBY181D. E. faecalis PBPX was used to exchange the terminal amino acid of stem peptides for biotin-D-Lys (BDL, yellow spheres), enabling visualization of glycan strands by western blotting with streptavidin. d, Glycan strands synthesized in vitro were treated with LytH. No difference in the distribution or intensity of polymers was observed between the three lanes. Blot is representative of two independent experiments. e, SDS-PAGE analysis of the co-immunoprecipitation experiment to identify proteins that bound to full-length LytH. Purple asterisks mark the protein bands corresponding to the baits. A red box is drawn around the ActH protein band. ActH is encoded by the gene saouhsc_01649. The fractions are: (FT) flowthrough, (W1-W2) washes, and (E1-E2) elutions. The schematics for LytH constructs are shown with the predicted transmembrane (TM), SH3, and amidase catalytic domains annotated. The predicted domain structure of ActH is also shown. f, A cladogram based on 16S rRNA alignment of bacterial species that encode ActH homologs with a similar domain structure was generated. The tree shows 196 bacterial species. For ease of visualization, only some species are labeled. Firmicutes are highlighted in purple and Deltaproteobacteria are highlighted in red.

Source data

Extended Data Fig. 4 The LytH-ActH complex has peptidoglycan hydrolase activity.

a, HG003 strains were spotted on TSA ± 0.125 μg/mL oxacillin. b, The relative ion count (extracted ion count/total ion count) for the tetrasaccharide-monopeptide was calculated. Data represent the mean ± SD. P-values were determined using unpaired, two-tailed t-tests (*P< 0.05; **P< 0.01; ***P< 0.001; ns, not significant). From left to right: P= 0.0048, 0.0005, 0.0387, 0.0010, 0.0580, and 0.0056. c, Full-length LytH and ActH were co-expressed in E. coli and purified from detergent-solubilized membranes as a stable complex. SDS- PAGE analysis of purification fractions: (FT) flowthrough. d, Western blot analysis of LytH reactions with glycan strand substrate, showing a distinct difference for the WT complex. Glycan strands were made in vitro from native S. aureus Lipid II; the strands were labeled with BDL for visualization. e, Western blot analysis of LytH reactions with glycan strand substrate, showing the catalytic serine of ActH is dispensable for activating LytH. The bold bands appearing in all lanes in the blot correspond to purified proteins that were added to the reactions and non-specifically labeled. Data are representative of three (b-c) and two (a, d-e) independent experiments.

Source data

Extended Data Fig. 5 The LytH-ActH complex removes stem peptides from uncrosslinked glycan strands.

a, Chemical structure highlighting the differences between synthetic and S. aureus native Lipid II. b, PAGE autoradiographs of reactions with uncrosslinked glycan strand substrates. The glycopolymers for substrates (1) and (3) were made from synthetic Lipid II, while the glycopolymers for substrate 2 were generated from native Lipid II. For substrate (1), the radiolabel was found in the [14C]-GlcNAc residues of the glycan backbone. For substrates (2) and (3), the radiolabel was provided by a short wall teichoic acid (WTA) disaccharide branch ([14C]-LIIAWTA) attached to the hydroxyl group at the C6 position. c, Purified S. aureus sacculi were treated with lysostaphin and LytHWT-ActH. Hydrolysis of sacculi was monitored over time as a decrease in OD600. Experiments were performed in triplicate for each concentration of protein; each line represents the mean, plotted as percent of initial absorbance. d, Schematic for a PAGE assay to assess if LytH-ActH cleaves crosslinked peptidoglycan. Uncrosslinked glycan strands (substrate 2) were made from native Lipid II and radiolabeled with a short [14C]-LIIAWTA disaccharide branch (substrate 1) at the C6 position. The radiolabeled glycan strands were crosslinked by S. aureus PBP4 to generate crosslinked peptidoglycan (substrate 3). e, Peptidoglycan substrates were treated with LytHWT-ActH or lysostaphin. Reaction products were resolved by PAGE. Lysostaphin cleaves crosslinks, generating smaller species that migrate faster in the gel. Molecular weight ladders were not run on these PAGE gels. Data are representative of three independent experiments (b-c, e).

Source data

Extended Data Fig. 6 Mass spectrometry confirms LytH-ActH amidase activity.

a, Glycan strands were made from native Lipid II using SgtBWT protein and treated with the LytH complex prior to LC-MS analysis. In the presence of LytHWT-ActH, the tetrasaccharide-monopeptide was observed, but only the monomer (starting material) was observed in the LytHD195A-ActH reaction. We could not detect the released stem peptide in these reactions with native glycan strands likely due to the poor ionization efficiency of the released pentaglycine-containing stem peptide. b, The chemical structure and isotope distributions for the disaccharide peptide species (blue peak) shown in Fig. 2f. c, The chemical structure and isotope distributions for the synthetic stem peptide product (red peak) shown in Fig. 2f.

Extended Data Fig. 7 PBP2 suppressor mutants showed reduced peptidoglycan synthesis activity.

a, Spot dilutions showing that excess PBP2 is toxic in the absence of LytH. Wild-type pbp2 (pbp2WT) or a pbp2* suppressor allele (pbp2N220→KDLN) was expressed from an inducible promoter in strain HG003 ΔlytH. In these strains, the native pbp2WT allele was still present. b, Fluorescence images of Nile red-stained PBP2 overexpression strains. The controls in which the inducer was withheld to prevent PBP2 overexpression are shown. Quantitation of cellular phenotypes is summarized in the graph below: cells with a nascent or complete septum at midcell (Class A), cells showing only membrane fluorescence (Class B), and cells showing fluorescent punctate foci or multiple septa (Class C). Scale bars, 1 µm. c, A PBP2 suppressor variant was purified. Bocillin- labeling of purified proteins showed that PBP2F158L was properly folded. The Coomassie gel was a control for amounts of protein loaded. The experiment was performed once. d, Western blot comparing PBP2WT and PBP2F158L reactions with Lipid II substrate. PBP2 polymerizes Lipid II into glycan strands and crosslinks the glycan strands; PBP2 also incorporates BDL to enable visualization of crosslinked peptidoglycan. The PBP2 reactions were incubated for 5 or 15 min. When indicated, lysostaphin was added to cleave peptidoglycan crosslinks so that highly-crosslinked material could enter into the gel. Data are representative of two independent experiments (a-b, d).

Source data

Extended Data Fig. 8 Reducing PBP2 activity suppresses lytH deletion defects.

a, Fluorescence images of Nile red-stained HG003 strains. The suppressor allele pbp2F158L (pbp2*) mitigates ∆lytH division defects. Quantitation of cellular phenotypes is shown in the graph to the right: cells with a nascent or complete septum at midcell (Class A), cells showing only membrane fluorescence (Class B), and cells showing multiple septa (Class C). Scale bars, 1 µm. b, Deletion of lytH leads to larger cell size. The distribution of cell volume was calculated for cells at different phases of growth: cells that have not initiated septal synthesis (phase 1), cells with a nascent septum (phase 2), and cells with a complete septum prior to splitting into two daughter cells (phase 3). Each dot represents a single cell (from left to right): n= 566, 516, 615, 259, 160, 201, 215, 145, and 241 cells. The median of each spread is indicated by a black line. P-values were determined by two-sided Mann-Whitney U tests (***P< 0.001). From left to right: P= 4.58x10-50, 8.10x10-86, 2.21x10-26, 1.94x10-30, 2.44x10-21, and 2.70x10-25. c, Transmission electron micrograph of the suppressor strain HG003 ∆lytH pbp2F158L is shown. Scale bar, 500 nm. The experiment was performed once at 30 °C and 37 °C. d, The number of cells with septal defects was counted. Class A cells showed normal placement of a nascent or complete septum at midcell, while Class B cells showed misplaced septa. Data are representative of two independent experiments (a-b).

Source data

Extended Data Fig. 9 Loss of LytH reduces peptidoglycan synthesis at the septum relative to the periphery.

a, Fluorescence images of fluorescein-D-lysine (FDL)-labeled cells. All FDL labeling was performed in strains lacking PBP4 to reduce background. Cells were sorted into different classes according to the pattern of FDL signal: at the septum (Class A), around the membrane (Class B), or at misplaced septa and/or peripheral puncta (Class C). Scale bars, 1 µm. b, The ratio of fluorescence intensity at the septum vs. periphery was calculated for cells with a single complete septum. Two regions of interests, outlined in red, were defined to retrieve pixel values at the periphery and the septum. c, Deletion of lytH leads to reduced septal to peripheral FDL signal independently of cell size. Each dot represents a single cell: n= 159 (wt) and 167 (∆lytH) cells. d, FDL fluorescence ratio is not correlated with cell size (n= 105 cells). Deletion of ezrA causes division defects and larger cell size. e, Fluorescence images of an FDL-labeled, mixed population of WT and ∆lytH cells. To distinguish WT from ∆lytH cells, one strain was stained with DAPI prior to mixing the two strains and labeling with FDL. Reciprocal labeling of DAPI was performed. Scale bars, 1 µm. f, FDL signal for mixed populations containing DAPI-stained WT cells. Fluorescence intensity measurements correspond to the median intensity at the periphery or septum. Different colored dots represent cells analyzed from different fields of view (sets 1-3): n= 52 (wt) and 17 (∆lytH) cells. P-values were determined by two-sided Mann-Whitney U tests (***P< 0.001; ns, not significant). From left to right: P= 0.9389 and 7.02x10-5. g, Fluorescence images of cells labelled with fluorescent vancomycin (Van-FL) to detect newly synthesized peptidoglycan. Scale bars, 1 μm. Data are representative of two independent experiments (a-g).

Source data

Extended Data Fig. 10 PBP2 was delocalized in the absence of LytH.

a, Fluorescence images of NCTC8325-4 strains expressing sGFP-PBP2. To compare the spatial distribution of PBP2 signal in WT vs. ∆lytH cells, fluorescence ratios were calculated using cells with a single complete septum. In all plots, each dot represents a single cell, the median of each spread is indicated by a black line, and p-values were determined by two-sided Mann-Whitney U tests. In this plot: n= 81 (wt) and 77 (∆lytH) cells; P= 8.30x10-8 (***P< 0.001). Scale bars, 1 µm. b, Fluorescence images of strain NCTC8325-4 ∆ezrA expressing sGFP-PBP2, showing that delocalization of PBP2 is independent of cell size. Fluorescence ratios of sGFP-PBP2 signal was plotted against cell volume (n= 91 cells). Scale bars, 2 µm. c, Fluorescence images of HG003 strains co-expressing sGFP-PBP2 and FtsZ-mCherry. PBP2 and FtsZ signals showed reduced colocalization in ∆lytH cells. Only cells showing FtsZ signal at the septum were considered for Pearson correlation coefficient analysis to calculate the degree of colocalization: n= 133 (wt) and 166 (∆lytH) cells; P= 0.006 (**P< 0.01). Scale bars, 1 µm. Different fields of view were captured for one biological sample (a-b). Data are representative of two (c) independent experiments.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–8 and Supplementary References.

Reporting Summary

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Fig. 3

Unprocessed western blots and/or gels.

Source Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 3

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 7

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 9

Statistical Source Data.

Source Data Extended Data Fig. 10

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Do, T., Schaefer, K., Santiago, A.G. et al. Staphylococcus aureus cell growth and division are regulated by an amidase that trims peptides from uncrosslinked peptidoglycan. Nat Microbiol 5, 291–303 (2020). https://doi.org/10.1038/s41564-019-0632-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0632-1

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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