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:

R-spondin-3 induces secretory, antimicrobial Lgr5+ cells in the stomach

Abstract

Wnt signalling stimulated by binding of R-spondin (Rspo) to Lgr-family members is crucial for gastrointestinal stem cell renewal. Infection of the stomach with Helicobacter pylori stimulates increased secretion of Rspo by myofibroblasts, leading to an increase in proliferation of Wnt-responsive Axin2+Lgr5 stem cells in the isthmus of the gastric gland and finally gastric gland hyperplasia. Basal Lgr5+ cells are also exposed to Rspo3, but their response remains unclear. Here, we demonstrate that—in contrast to its known mitogenic activity—Rspo3 induces differentiation of basal Lgr5+ cells into secretory cells that express and secrete antimicrobial factors, such as intelectin-1, into the lumen. The depletion of Lgr5+ cells or the knockout of Rspo3 in myofibroblasts leads to hypercolonization of the gastric glands with H. pylori, including the stem cell compartment. By contrast, systemic administration or overexpression of Rspo3 in the stroma clears H. pylori from the gastric glands. Thus, the Rspo3–Lgr5 axis simultaneously regulates both antimicrobial defence and mucosal regeneration.

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: Endogenous Rspo3 controls antral-gland proliferation and stem-cell signature.
Fig. 2: Rspo3 controls the secretory phenotype in the base of antrum glands.
Fig. 3: A subpopulation of Lgr5+ cells expresses antimicrobial genes to counterbalance infection.
Fig. 4: Gene expression and morphology of organoids grown with different concentrations of Rspo.
Fig. 5: Rspo3 counterbalances H. pylori infection.
Fig. 6: Overexpression of Rspo3 in myofibroblasts expands the gland base mucous cells and limits gland colonization.
Fig. 7: After infection, Lgr5+ cells secrete Itln1 into the gland lumen to bind and agglutinate H. pylori.

Similar content being viewed by others

Data availability

The microarray data from this manuscript have been deposited in the GEO under accession code GSE108729. Single-cell RNA-seq data from this manuscript have been deposited in the GEO under accession code GSE124930. Bulk RNA-seq data are available from GEO under accession code GSE129107. Quantitative data that support the findings of this study are available within the paper and the Supplementary Information. Source data for graphical representations and statistical descriptions have been provided as Supplementary Table 6. All other data that support the findings of this study are available from the corresponding author on reasonable request.

Code availability

The computational code for GSEA analysis of microarray data of this manuscript can be accessed at https://github.com/MPIIB-Department-TFMeyer/Sigal_et_al_Rspo3_secretory_role. The code used for single cell RNA-seq analysis can be accessed at https://gist.github.com/cofischer/ab41169b5212046a59ba831e280835b3.

References

  1. Parsonnet, J. et al. Helicobacter pylori infection and the risk of gastric carcinoma. N. Engl. J. Med. 325, 1127–1131 (1991).

    Article  CAS  Google Scholar 

  2. Amieva, M. R. et al. Disruption of the epithelial apical-junctional complex by Helicobacter pylori CagA. Science 300, 1430–1434 (2003).

    Article  CAS  Google Scholar 

  3. Toller, I. M. et al. Carcinogenic bacterial pathogen Helicobacter pylori triggers DNA double-strand breaks and a DNA damage response in its host cells. Proc. Natl Acad. Sci. USA 108, 14944–14949 (2011).

    Article  CAS  Google Scholar 

  4. Sigal, M. et al. Helicobacter pylori activates and expands Lgr5(+) stem cells through direct colonization of the gastric glands. Gastroenterology 148, 1392–404.e21 (2015).

    Article  CAS  Google Scholar 

  5. Sigal, M. et al. Stromal R-spondin orchestrates gastric epithelial stem cells and gland homeostasis. Nature 548, 451–455 (2017).

    Article  CAS  Google Scholar 

  6. Munoz, J. et al. The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+4’ cell markers. EMBO J. 31, 3079–3091 (2012).

    Article  CAS  Google Scholar 

  7. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  Google Scholar 

  8. Arnold, I. et al. Tolerance rather than immunity protects from Helicobacter pylori-induced gastric preneoplasia. Gastroenterology 140, 199–209 (2010).

    Article  Google Scholar 

  9. Wunder, C. et al. Cholesterol glucosylation promotes immune evasion by Helicobacter pylori. Nat. Med. 12, 1030–1038 (2006).

    Article  CAS  Google Scholar 

  10. Komiya, T., Tanigawa, Y. & Hirohashi, S. Cloning of the novel gene intelectin, which is expressed in intestinal Paneth cells in mice. Biochem. Biophys. Res. Commun. 251, 759–762 (1998).

    Article  CAS  Google Scholar 

  11. Wesener, D. A. et al. Recognition of microbial glycans by human intelectin-1. Nat. Struct. Mol. Biol. 22, 603–610 (2015).

    Article  CAS  Google Scholar 

  12. Powell, J. J., Greenfield, S. M. & Thompson, R. P. Concentrations of metals in gastric juice in health and peptic ulcer disease. Gut 33, 1617–1620 (1992).

    Article  CAS  Google Scholar 

  13. Stange, D. E. et al. Differentiated Troy + chief cells act as reserve stem cells to generate all lineages of the stomach epithelium. Cell 155, 357–368 (2013).

    Article  CAS  Google Scholar 

  14. Leushacke, M. et al. Lgr5-expressing chief cells drive epithelial regeneration and cancer in the oxyntic stomach. Nat. Cell Biol. 19, 774–786 (2017).

    Article  CAS  Google Scholar 

  15. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  Google Scholar 

  16. Farin, H. F. et al. Visualization of a short-range Wnt gradient in the intestinal stem-cell niche. Nature 530, 340–343 (2016).

    Article  CAS  Google Scholar 

  17. Wehkamp, J. et al. Paneth cell antimicrobial peptides: topographical distribution and quantification in human gastrointestinal tissues. FEBS Lett. 580, 5344–5350 (2006).

    Article  CAS  Google Scholar 

  18. Koslowski, M. J. et al. Association of a functional variant in the Wnt co-receptor LRP6 with early onset ileal Crohn’s disease. PLoS Genet. 8, e1002523 (2012).

    Article  CAS  Google Scholar 

  19. Saenz, J. B., Vargas, N. & Mills, J. C. Tropism for spasmolytic polypeptide-expressing metaplasia allows Helicobacter pylori to expand its intragastric niche. Gastroenterology 156, 160–174 (2019).

    Article  Google Scholar 

  20. Naik, S., Larsen, S. B., Cowley, C. J. & Fuchs, E. Two to tango: dialog between immunity and stem cells in health and disease. Cell 175, 908–920 (2018).

    Article  CAS  Google Scholar 

  21. Bartfeld, S. et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection.Gastroenterology 148, 126–136.e6 (2015).

    Article  Google Scholar 

  22. Boccellato, F. et al. Polarised epithelial monolayers of the gastric mucosa reveal insights into mucosal homeostasis and defence against infection. Gut 68, 400–413 (2019).

    Article  CAS  Google Scholar 

  23. Zimmermann, S. et al. ALPK1- and TIFA-dependent innate immune response triggered by the Helicobacter pylori type IV secretion system. Cell Rep. 20, 2384–2395 (2017).

    Article  CAS  Google Scholar 

  24. Pfannkuch, L. et al. ADP heptose, a novel pathogen-associated molecular pattern identified in Helicobacter pylori. FASEB J. https://doi.org/10.1096/fj.201802555R (2019).

  25. Arnold, I. C. et al. Tolerance rather than immunity protects from Helicobacter pylori-induced gastric preneoplasia. Gastroenterology 140, 199–209 (2011).

    Article  CAS  Google Scholar 

  26. Mejias-Luque, R. et al. Lymphotoxin β receptor signalling executes Helicobacter pylori-driven gastric inflammation in a T4SS-dependent manner. Gut 66, 1369–1381 (2017).

    Article  CAS  Google Scholar 

  27. Amieva, M. R. & El-Omar, E. M. Host-bacterial interactions in Helicobacter pylori infection. Gastroenterology 134, 306–323 (2008).

    Article  CAS  Google Scholar 

  28. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011).

  29. Neufeld, S. et al. A conditional allele of Rspo3 reveals redundant function of R-spondins during mouse limb development. Genesis 50, 741–749 (2012).

    Article  CAS  Google Scholar 

  30. Herring, B. P., Hoggatt, A. M., Burlak, C. & Offermanns, S. Previously differentiated medial vascular smooth muscle cells contribute to neointima formation following vascular injury. Vasc. Cell 6, 21 (2014).

    Article  Google Scholar 

  31. Hilkens, J. et al. RSPO3 expands intestinal stem cell and niche compartments and drives tumorigenesis. Gut 66, 1095–1105 (2017).

    Article  CAS  Google Scholar 

  32. Dyer, V. et al. Genomic features of the Helicobacter pylori strain PMSS1 and its virulence attributes as deduced from its in vivo colonisation patterns. Mol. Microbiol. 110, 761–776 (2018).

    Article  CAS  Google Scholar 

  33. Churchill, G. A. Fundamentals of experimental design for cDNA microarrays. Nat. Genet. 32, 490–495 (2002).

    Article  CAS  Google Scholar 

  34. Yu, D., Huber, W. & Vitek, O. Shrinkage estimation of dispersion in negative binomial models for RNA-seq experiments with small sample size. Bioinformatics 29, 1275–1282 (2013).

    Article  CAS  Google Scholar 

  35. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  Google Scholar 

  36. Kamburov, A., Stelzl, U., Lehrach, H. & Herwig, R. The ConsensusPathDB interaction database: 2013 update. Nucleic Acids Res. 41, D793–D800 (2013).

    Article  CAS  Google Scholar 

  37. Sergushichev, A. An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. Preprint at bioRxiv https://doi.org/10.1101/060012 (2016).

  38. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  39. Backert, S. et al. Translocation of the Helicobacter pylori CagA protein in gastric epithelial cells by a type IV secretion apparatus. Cell. Microbiol. 2, 155–164 (2000).

    Article  CAS  Google Scholar 

  40. Koch, M., Mollenkopf, H. J. & Meyer, T. F. Macrophages recognize the Helicobacter pylori type IV secretion system in the absence of toll-like receptor signalling. Cell. Microbiol. 18, 137–147 (2016).

    Article  CAS  Google Scholar 

  41. Hatzios, S. K. et al. Chemoproteomic profiling of host and pathogen enzymes active in cholera. Nat. Chem. Biol. 12, 268–274 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Hoffmann, S. Lehmann, Y. Giesecke, I. Wagner and C. Braeuning for technical support, V. Brinkmann for help with live imaging, A. Kühl and members of the Charité Core Facility for Histopathology for sample processing and histopathological scoring, and R. Zietlow for editing the manuscript. This work was performed in the Department of Molecular Biology of the MPIIB with extramural support from the DFG to M.S. (SI1983/2-1 and SI1983/3-1) and T.F.M. (ME705/18-1). M.S. received financial support from the George-Will-Foundation and is a participant of the BIH Charité Clinician Scientist Program funded by the Charité University Medicine Berlin and the Berlin Institute of Health.

Author information

Authors and Affiliations

Authors

Contributions

The study was designed by M.S. and supervised by T.F.M. M.S. and S.M. performed most of the in vivo experiments. M.E.R. and M.S. performed Lgr5 cell sorting and RNA bulk sequencing experiments. M.d.M.R. performed the functional experiments with Itln1. M.K., C.F. and S.S. performed single-cell RNA-seq experiments and data analysis. H.-J.M. performed microarray studies and H.B. and H.-J.M. analysed the data. E.R.M.B. and B.W. contributed materials required for the study. The manuscript was written by M.S. with guidance and discussion from T.F.M.

Corresponding authors

Correspondence to Michael Sigal or Thomas F. Meyer.

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.

Integrated supplementary information

Supplementary Fig. 1 R-spondin 3 affects Lgr5 signature and colonization with H. pylori.

a) Single molecule RNA-in situ hybridization (red dots) for Rspo3 in corpus tissue. b) GSEA from microarray data comparing the expression profile of corpus tissue of Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice treated with tamoxifen 14 days before sacrifice to published intestinal Lgr5 signatures and TROY+ cell signature in the corpus. A permutation test was used. p-values from all gene sets were jointly adjusted for multiple testing using the method of Benjamini-Hochberg (FDR). c) qPCR analysis of Lgr5 expression normalized to Gapdh from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice. d) H&E images from the corpus of Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice 2 months before sacrifice. e) Immunofluorescence images of stomach antrum tissue from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with H. pylori (green) 2 months before sacrifice. f) Expression levels of genes encoding for Reg proteins in the corpus of 2-month-infected Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice. Data from n = 2 independent biological samples per group. g) qPCR for Reg3g expression normalized to Gapdh in the corpus of uninfected and 2-month-infected Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice. Data are displayed as mean±SD from 3 biological replicates, analyzed by two-sided Student’s t-test. Images are representative of at least 3 independent biological samples. Scale bars: 100 μm. Microarray data are from 2 independent biological samples per group.

Supplementary Fig. 2 Bulk RNAseq from Lgr5+ cells.

a) Schematic visualization of a gland with Lgr5eGFP+ cells in the base b) Table of genes with highest FPKM values in uninfected Lgr5+ cells (data from n = 3 independent biological samples) c) FPKM values for EPCAM and Lgr5 in uninfected and infected samples. d) FPKM values of TFF2 as a marker of basal cells and Muc5ac as a marker of surface pit cells. e) Gating and sorting strategy of Lgr5+ EPCAM+ cells (note that all Lgr5+ cells are EPCAM+). Data are displayed as mean ± SD from 3 independent biological samples

Supplementary Fig. 3 Acidity is not affected by Rspo3.

a) Immunofluorescence for gastrin in mouse antrum from uninfected and 2-month-infected Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice. b) Expression of gastrin from microarray data comparing the transcriptome of Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mouse antrum (mice were sacrificed 14 d post tamoxifen). c) Relative expression of gastrin mRNA normalized to Gapdh in the antrum of WT and Rspo3 KO mice either uninfected (n = 3 and n = 4) or infected with H. pylori for 2 months (n = 3 and n = 4). d) Relative expression of Atp4b mRNA normalized to Gapdh in the corpus of WT and Rspo3 KO mice either uninfected (n = 3 and n = 4) or infected with H. pylori for 2 months (n = 3 and n = 4). e) pH measured in the stomachs of WT and Rspo3 KO mice either uninfected (n = 4 and n = 5) or infected with H. pylori for 2 months (n = 5 and n = 4). Data are displayed as mean ± SD, analyzed by one-way ANOVA. Images are representative of at least 3 independent biological samples. Scale bars: 100 μm.

Supplementary Fig. 4 Expression of Lgr5 and intelectin is Rspo3-dependent, but overexpression is not observed early (2 weeks) after infection.

a) qPCR data from uninfected (n = 3) and 2 week-infected (n = 4) WT mice relative to Gapdh, normalized to uninfected control. b) qPCR data from 2 week-infected Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice (n = 4 per group) relative to Gapdh, normalized to Myh11CreErt2/Rspo3+/+. Data are displayed as mean ± SD, analyzed by two-sided Student’s t-test.

Supplementary Fig. 5 Functional T4SS and Rspo3 are required for antimicrobial effects but Rspo3 does not affect the inflammatory response.

a) Quantification of colony forming units from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with WT or ΔCagE H. pylori strain PMSS1 and treated with tamoxifen 2 months before sacrifice (for WT PMSS1 infection, data from Fig. 3a is reproduced here), each dot represents one mouse (n = 6,7,3 and 6 per group as indicated in the figure). b) qPCR for Lgr5 expression normalized to Gapdh from mouse antrum of Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl infected with WT and ΔCagE PMSS1 strain of H. pylori 2 months before sacrifice (n = 5,3,3,5 per group). c) qPCR for Itln1 expression normalized to Gapdh from antrum of Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice (n = 5,3,3,5 per respective group as indicated in the figure). d) Inflammation score for uninfected mice and Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice (n = 7,6,3,4 per respective group as indicated in the figure). e) Quantification of F4/80 immunostaining from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice (data from 3 mice per group, comparison between Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl for each pair (uninfected, WT infected, ΔCagE infected) was done by two-sided t test). f) Quantification of CD3 immunostaining from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice (data from 3 mice per group, comparison between Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl for each pair (uninfected, WT infected, ΔCagE infected) was done using two-sided t test). g) Representative images for F4/80 immunostaining from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice. Images representative of 3 independent biological samples per group. h) Representative images for CD3 immunostaining from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice. Images representative of 3 independent biological samples per group. i) Quantification of CD4+ T-bet+ cells measured in 10 high power fields per mouse from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl infected with WT and ΔCagE H. pylori strain PMSS1 2 months before sacrifice (n = 4,3,4,5 per respective group), one way ANOVA. j) qPCR for interferon-γ and CXCL9 normalized to Gapdh from Myh11CreErt2/Rspo3+/+ and Myh11CreErt2/Rspo3fl/fl mice infected with WT H. pylori for 2 months, n = 4 per group, two-sided t-test. Scale bars: 100 µm.

Supplementary Fig. 6 Intelectin expression is Rspo-dependent in organoids and restricted to the gland base.

a-c) qPCR for Lgr5, Axin2 and Itln1 in primary 3D organoids derived from the mouse stomach antrum and grown in medium supplemented with different concentrations of Rspo1, normalized to Gapdh (0 and 50%: n = 2 independent biological replicates, 100%: n = 3 independent biological samples). d) smFISH for Itln1 in the antrum of an uninfected and a 2-month-infected mouse. Images are representative of 3 independent biological samples. Scale bars: 100 μm. e) Histogram displaying the position of Itln1+ cells in the antrum glands in 2-month-infected mice, data derived from n = 3 mice. f) Staining for Ki67, as well as high magnification image of co-staining for GSII and Ki67 to demonstrate that the gland base contains mainly GSII+ cells, whereas proliferation is mainly present in the cells above the base. Immunofluorescence for CD44 demonstrates expression in the gland base. Data are displayed as mean ± SD. Images are representative of at least 3 independent biological samples. Scale bars: 100 μm.

Supplementary Fig. 7 Intelectin 1 agglutinates H. pylori.

a) Survival curves: CFU data upon exposure of H. pylori cultures to Itln1, HBD3 and polymyxin B at different concentrations. Data derived from n = 3 independent biological samples. b) Agglutination assay: Crystal violet images of H. pylori cultures exposed to polymyxin B right: Quantification from n = 3 independent biological samples per group. c) Combined confocal microscopy/phase contrast images of H. pylori cultures either exposed to recombinant Itln1 (right) or the same volume of PBS. The cultures were recorded for a total of 52 minutes and images are from minute 26. The experiment was repeated twice in the lab. The area of GFP+ clusters that exceeded the size of single bacteria was measured every 60 seconds and plotted as GFP+ area over time. Red dots represent Itln1-treated cultures; black dots represent PBS-treated controls. d) Confocal microscopy images of primary bone marrow-derived macrophages that were infected with H. pylori in the presence or absence of recombinant Itln1. Middle: Inserts from the images at higher magnification. The GFP+ area per macrophage (n = 51 and n = 52 per group) was measured and two sided t-test was performed. Data are displayed as mean±SD. Scale bars: 10 µm.

Supplementary Fig. 8

Unprocessed blot of Fig. 7c

Supplementary Information

Supplementary Information

Supplementary Figs. 1–8

Reporting Summary

Supplementary Table 1

FPKM values from bulk sequenced Lgr5eGFP+ cells

Supplementary Table 2

Top 50 cluster specific genes for each cluster from scRNAseq data from LgreGFP+ cells of uninfected and H. pylori infected mice.

Supplementary Table 3

Gene sets of differentially expressed genes between cluster 1 and non-cluster 1 genes from data 18 of infected cells were subjected to over-representation analysis using the web-based tool 19 ConsensusPathDB-mouse.

Supplementary Table 4

Differential gene expression analysis from cluster 1 and cluster 2 from uninfected and infected cells using the merged dataset from single cells RNAsec (negative binomial exact test).

Supplementary Table 5

Summary of technical and statistical information from single cell RNAseq experiment.

Supplementary Table 6

Statistics source data

Supplementary Video 1

Time lapse movie (total of 50 minutes) displaying bacterial clustering upon exposure to intelectin 1.

Supplementary Video 2

Real-time visualization of bacterial motility from movie 1 to visualize altered bacterial motility upon intelectin 1 treatment.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sigal, M., Reinés, M.d.M., Müllerke, S. et al. R-spondin-3 induces secretory, antimicrobial Lgr5+ cells in the stomach. Nat Cell Biol 21, 812–823 (2019). https://doi.org/10.1038/s41556-019-0339-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0339-9

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