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:

CYRI/FAM49B negatively regulates RAC1-driven cytoskeletal remodelling and protects against bacterial infection

Abstract

Salmonella presents a global public health concern. Central to Salmonella pathogenicity is an ability to subvert host defences through strategically targeting host proteins implicated in restricting infection. Therefore, to gain insight into the host–pathogen interactions governing Salmonella infection, we performed an in vivo genome-wide mutagenesis screen to uncover key host defence proteins. This revealed an uncharacterized role of CYRI (FAM49B) in conferring host resistance to Salmonella infection. We show that CYRI binds to the small GTPase RAC1 through a conserved domain present in CYFIP proteins, which are known RAC1 effectors that stimulate actin polymerization. However, unlike CYFIP proteins, CYRI negatively regulates RAC1 signalling, thereby attenuating processes such as macropinocytosis, phagocytosis and cell migration. This enables CYRI to counteract Salmonella at various stages of infection, including bacterial entry into non-phagocytic and phagocytic cells as well as phagocyte-mediated bacterial dissemination. Intriguingly, to dampen its effects, the bacterial effector SopE, a RAC1 activator, selectively targets CYRI following infection. Together, this outlines an intricate host–pathogen signalling interplay that is crucial for determining bacterial fate. Notably, our study also outlines a role for CYRI in restricting infection mediated by Mycobacterium tuberculosis and Listeria monocytogenes. This provides evidence implicating CYRI cellular functions in host defence beyond Salmonella infection.

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: The Ity15 pedigree displays increased susceptibility to S. Typhimurium infection.
Fig. 2: A mutation in Cyri confers susceptibility to S. Typhimurium infection in the Ity15 pedigree.
Fig. 3: CYRI binds to the small GTPase RAC1 in a GTP-dependent manner.
Fig. 4: CYRI regulates cell morphology and actin cytoskeletal dynamics in a RAC1-dependent manner.
Fig. 5: CYRI negatively regulates SopE-mediated cellular effects.
Fig. 6: CYRI expression in haematopoietic cells confers resistance to S. Typhimurium infection.
Fig. 7: The Salmonella bacterial effector SopE negatively regulates CYRI.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors on reasonable request.

Code availability

All commercial and custom codes used to generate data presented in this study are available from the corresponding authors on reasonable request.

References

  1. Kirk, M. D. et al. World Health Organization estimates of the global and regional disease burden of 22 foodborne bacterial, protozoal, and viral diseases, 2010: a data synthesis. PLoS Med. 12, e1001921 (2015).

    Article  Google Scholar 

  2. Majowicz, S. E. et al. The global burden of nontyphoidal Salmonella gastroenteritis. Clin. Infect. Dis. 50, 882–889 (2010).

    Article  Google Scholar 

  3. Prestinaci, F., Pezzotti, P. & Pantosti, A. Antimicrobial resistance: a global multifaceted phenomenon. Pathog. Glob. Health 109, 309–318 (2015).

    Article  Google Scholar 

  4. LaRock, D. L., Chaudhary, A. & Miller, S. I. Salmonellae interactions with host processes. Nat. Rev. Microbiol. 13, 191–205 (2015).

    Article  CAS  Google Scholar 

  5. Schlumberger, M. C. & Hardt, W. D. Salmonella type III secretion effectors: pulling the host cell’s strings. Curr. Opin. Microbiol. 9, 46–54 (2006).

    Article  CAS  Google Scholar 

  6. Byndloss, M. X., Rivera-Chavez, F., Tsolis, R. M. & Baumler, A. J. How bacterial pathogens use type III and type IV secretion systems to facilitate their transmission. Curr. Opin. Microbiol. 35, 1–7 (2017).

    Article  CAS  Google Scholar 

  7. Guiney, D. G. & Lesnick, M. Targeting of the actin cytoskeleton during infection by Salmonella strains. Clin. Immunol. 114, 248–255 (2005).

    Article  CAS  Google Scholar 

  8. Hardt, W. D., Chen, L. M., Schuebel, K. E., Bustelo, X. R. & Galan, J. E. S. typhimurium encodes an activator of Rho GTPases that induces membrane ruffling and nuclear responses in host cells. Cell 93, 815–826 (1998).

    Article  CAS  Google Scholar 

  9. Orchard, R. C. & Alto, N. M. Mimicking GEFs: a common theme for bacterial pathogens. Cell. Microbiol. 14, 10–18 (2012).

    Article  CAS  Google Scholar 

  10. Humphreys, D., Davidson, A., Hume, P. J. & Koronakis, V. Salmonella virulence effector SopE and host GEF ARNO cooperate to recruit and activate WAVE to trigger bacterial invasion. Cell Host Microbe 11, 129–139 (2012).

    Article  CAS  Google Scholar 

  11. Gautreau, A. et al. Purification and architecture of the ubiquitous Wave complex. Proc. Natl Acad. Sci. USA 101, 4379–4383 (2004).

    Article  CAS  Google Scholar 

  12. Eden, S., Rohatgi, R., Podtelejnikov, A. V., Mann, M. & Kirschner, M. W. Mechanism of regulation of WAVE1-induced actin nucleation by Rac1 and Nck. Nature 418, 790–793 (2002).

    Article  CAS  Google Scholar 

  13. Ismail, A. M., Padrick, S. B., Chen, B., Umetani, J. & Rosen, M. K. The WAVE regulatory complex is inhibited. Nat. Struct. Mol. Biol. 16, 561–563 (2009).

    Article  CAS  Google Scholar 

  14. Chen, Z. et al. Structure and control of the actin regulatory WAVE complex. Nature 468, 533–538 (2010).

    Article  CAS  Google Scholar 

  15. Chen, B. et al. Rac1 GTPase activates the WAVE regulatory complex through two distinct binding sites. eLife 6, e29795 (2017).

    Article  Google Scholar 

  16. Francis, C. L., Ryan, T. A., Jones, B. D., Smith, S. J. & Falkow, S. Ruffles induced by Salmonella and other stimuli direct macropinocytosis of bacteria. Nature 364, 639–642 (1993).

    Article  CAS  Google Scholar 

  17. Steele-Mortimer, O. The Salmonella-containing vacuole: moving with the times. Curr. Opin. Microbiol. 11, 38–45 (2008).

    Article  CAS  Google Scholar 

  18. Ham, H., Sreelatha, A. & Orth, K. Manipulation of host membranes by bacterial effectors. Nat. Rev. Microbiol. 9, 635–646 (2011).

    Article  CAS  Google Scholar 

  19. Schleker, S. et al. The current Salmonella-host interactome. Proteom. Clin. Appl. 6, 117–133 (2012).

    Article  CAS  Google Scholar 

  20. Mostowy, S. & Shenoy, A. R. The cytoskeleton in cell-autonomous immunity: structural determinants of host defence. Nat. Rev. Immunol. 15, 559–573 (2015).

    Article  CAS  Google Scholar 

  21. Behnsen, J., Perez-Lopez, A., Nuccio, S. P. & Raffatellu, M. Exploiting host immunity: the Salmonella paradigm. Trends Immunol. 36, 112–120 (2015).

    Article  CAS  Google Scholar 

  22. Worley, M. J., Nieman, G. S., Geddes, K. & Heffron, F. Salmonella typhimurium disseminates within its host by manipulating the motility of infected cells. Proc. Natl Acad. Sci. USA 103, 17915–17920 (2006).

    Article  CAS  Google Scholar 

  23. Ohl, M. E. & Miller, S. I. Salmonella: a model for bacterial pathogenesis. Annu. Rev. Med. 52, 259–274 (2001).

    Article  CAS  Google Scholar 

  24. Fabrega, A. & Vila, J. Salmonella enterica serovar Typhimurium skills to succeed in the host: virulence and regulation. Clin. Microbiol. Rev. 26, 308–341 (2013).

    Article  CAS  Google Scholar 

  25. Vazquez-Torres, A. et al. Extraintestinal dissemination of Salmonella by CD18-expressing phagocytes. Nature 401, 804–808 (1999).

    Article  CAS  Google Scholar 

  26. Hendriksen, R. S. et al. Global monitoring of Salmonella serovar distribution from the world health organization global foodborne infections network country data bank: results of quality assured laboratories from 2001 to 2007. Foodborne Pathog. Dis. 8, 887–900 (2011).

    Article  Google Scholar 

  27. Soding, J., Biegert, A. & Lupas, A. N. The HHpred interactive server for protein homology detection and structure prediction. Nucleic Acids Res. 33, W244–W248 (2005).

    Article  Google Scholar 

  28. Michiels, F., Habets, G. G., Stam, J. C., van der Kammen, R. A. & Collard, J. G. A role for Rac in Tiam1-induced membrane ruffling and invasion. Nature 375, 338–340 (1995).

    Article  CAS  Google Scholar 

  29. Krauthammer, M. et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat. Genet. 44, 1006–1014 (2012).

    Article  CAS  Google Scholar 

  30. Pollard, T. D. & Cooper, J. A. Actin, a central player in cell shape and movement. Science 326, 1208–1212 (2009).

    Article  CAS  Google Scholar 

  31. Spector, I., Shochet, N. R., Kashman, Y. & Groweiss, A. Latrunculins: novel marine toxins that disrupt microfilament organization in cultured cells. Science 219, 493–495 (1983).

    Article  CAS  Google Scholar 

  32. May, R. C. & Machesky, L. M. Phagocytosis and the actin cytoskeleton. J. Cell Sci. 114, 1061–1077 (2001).

    CAS  PubMed  Google Scholar 

  33. Krause, M. & Gautreau, A. Steering cell migration: lamellipodium dynamics and the regulation of directional persistence. Nat. Rev. Mol. Cell Biol. 15, 577–590 (2014).

    Article  CAS  Google Scholar 

  34. Wijburg, O. L., Simmons, C. P., van Rooijen, N. & Strugnell, R. A. Dual role for macrophages in vivo in pathogenesis and control of murine Salmonella enterica var. Typhimurium infections. Eur. J. Immunol. 30, 944–953 (2000).

    Article  CAS  Google Scholar 

  35. Richter-Dahlfors, A., Buchan, A. M. & Finlay, B. B. Murine salmonellosis studied by confocal microscopy: Salmonella typhimurium resides intracellularly inside macrophages and exerts a cytotoxic effect on phagocytes in vivo. J. Exp. Med. 186, 569–580 (1997).

    Article  CAS  Google Scholar 

  36. Lin, D. C. & Lin, S. Actin polymerization induced by a motility-related high-affinity cytochalasin binding complex from human erythrocyte membrane. Proc. Natl Acad. Sci. USA 76, 2345–2349 (1979).

    Article  CAS  Google Scholar 

  37. Campa, C. C. et al. Rac signal adaptation controls neutrophil mobilization from the bone marrow. Sci. Signal. 9, ra124 (2016).

    Article  Google Scholar 

  38. Pankov, R. et al. A Rac switch regulates random versus directionally persistent cell migration. J. Cell Biol. 170, 793–802 (2005).

    Article  CAS  Google Scholar 

  39. Schlumberger, M. C. et al. Amino acids of the bacterial toxin SopE involved in G nucleotide exchange on Cdc42. J. Biol. Chem. 278, 27149–27159 (2003).

    Article  CAS  Google Scholar 

  40. Fiskin, E., Bionda, T., Dikic, I. & Behrends, C. Global analysis of host and bacterial ubiquitinome in response to Salmonella Typhimurium infection. Mol. Cell 62, 967–981 (2016).

    Article  CAS  Google Scholar 

  41. de Chastellier, C. & Berche, P. Fate of Listeria monocytogenes in murine macrophages: evidence for simultaneous killing and survival of intracellular bacteria. Infect. Immun. 62, 543–553 (1994).

    PubMed  PubMed Central  Google Scholar 

  42. Cossart, P. & Lebreton, A. A trip in the “New Microbiology” with the bacterial pathogen Listeria monocytogenes. FEBS Lett. 588, 2437–2445 (2014).

    Article  CAS  Google Scholar 

  43. Rajaram, M. V. S. et al. M. tuberculosis-initiated human mannose receptor signaling regulates macrophage recognition and vesicle trafficking by FcRγ-Chain, Grb2, and SHP-1. Cell Rep. 21, 126–140 (2017).

    Article  CAS  Google Scholar 

  44. Ireton, K., Rigano, L. A. & Dowd, G. C. Role of host GTPases in infection by Listeria monocytogenes. Cell Microbiol. 16, 1311–1320 (2014).

    Article  CAS  Google Scholar 

  45. Shang, W. et al. Genome-wide CRISPR screen identifies FAM49B as a key regulator of actin dynamics and T cell activation. Proc. Natl Acad. Sci. USA 115, E4051–E4060 (2018).

    Article  CAS  Google Scholar 

  46. Fort, L. et al. Fam49/CYRI interacts with Rac1 and locally suppresses protrusions. Nat. Cell Biol. 20, 1159–1171 (2018).

    Article  CAS  Google Scholar 

  47. Queval, C. J., Brosch, R. & Simeone, R. The macrophage: a disputed fortress in the battle against Mycobacterium tuberculosis. Front. Microbiol. 8, 2284 (2017).

    Article  Google Scholar 

  48. Haney, M. S. et al. Identification of phagocytosis regulators using magnetic genome-wide CRISPR screens. Nat. Genet. 50, 1716–1727 (2018).

    Article  CAS  Google Scholar 

  49. Drevets, D. A. Dissemination of Listeria monocytogenes by infected phagocytes. Infect. Immun. 67, 3512–3517 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Maculins, T., Fiskin, E., Bhogaraju, S. & Dikic, I. Bacteria-host relationship: ubiquitin ligases as weapons of invasion. Cell Res. 26, 499–510 (2016).

  51. Roy, M. F. et al. Pyruvate kinase deficiency confers susceptibility to Salmonella typhimurium infection in mice. J. Exp. Med. 204, 2949–2961 (2007).

    Article  CAS  Google Scholar 

  52. Barthel, M. et al. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar Typhimurium colitis model that allows analysis of both pathogen and host. Infect. Immun. 71, 2839–2858 (2003).

    Article  CAS  Google Scholar 

  53. Eva, M. M. et al. Altered IFN-γ-mediated immunity and transcriptional expression patterns in N-ethyl-N-nitrosourea-induced STAT4 mutants confer susceptibility to acute typhoid-like disease. J. Immunol. 192, 259–270 (2014).

    Article  CAS  Google Scholar 

  54. Marei, H. et al. Differential Rac1 signalling by guanine nucleotide exchange factors implicates FLII in regulating Rac1-driven cell migration. Nat. Commun. 7, 10664 (2016).

    Article  CAS  Google Scholar 

  55. Benard, V., Bohl, B. P. & Bokoch, G. M. Characterization of Rac and Cdc42 activation in chemoattractant-stimulated human neutrophils using a novel assay for active GTPases. J. Biol. Chem. 274, 13198–13204 (1999).

    Article  CAS  Google Scholar 

  56. Marei, H., Carpy, A., Macek, B. & Malliri, A. Proteomic analysis of Rac1 signaling regulation by guanine nucleotide exchange factors. Cell Cycle 15, 1961–1974 (2016).

    Article  CAS  Google Scholar 

  57. Karlinsey, J. E. λ-Red genetic engineering in Salmonella enterica serovar Typhimurium. Methods Enzym. 421, 199–209 (2007).

    Article  CAS  Google Scholar 

  58. Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011).

    Article  CAS  Google Scholar 

  59. Fortin, A. et al. Recombinant congenic strains derived from A/J and C57BL/6J: a tool for genetic dissection of complex traits. Genomics 74, 21–35 (2001).

    Article  CAS  Google Scholar 

  60. Cunnington, A. J., de Souza, J. B., Walther, M. & Riley, E. M. Malaria impairs resistance to Salmonella through heme- and heme oxygenase-dependent dysfunctional granulocyte mobilization. Nat. Med. 18, 120–127 (2011).

    Article  Google Scholar 

  61. Sixt, M. & Lammermann, T. In vitro analysis of chemotactic leukocyte migration in 3D environments. Methods Mol. Biol. 769, 149–165 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank L. Larivière, E. Flamant, L. Rached-D’Astous, J. Kim, F. Pampaloni and M. Sader for technical assistance, N. Prud’homme and P. D’Arcy for mouse breeding and screening, S.-J. Pilon for histopathology scoring, and K. Koch and T. Maculins for reviewing the manuscript. We also acknowledge A. Malliri for reagents and G. White for the preparation of the PAK-CRIB peptide. This work was supported by the Canadian Institutes of Health Research (grant no. MOP133700 to D.M.), the DFG-funded Collaborative Research Centre on Selective Autophagy (grant no. SFB 1177), the European Research Council under the European Union’s Horizon 2020 research and innovation program (grant agreement no. 742720), the DFG-funded Cluster of Excellence ‘Macromolecular Complexes’ (grant no. EXC115), the DFG-funded SPP 1580 program ‘Intracellular Compartments as Places of Pathogen-Host-Interactions’, and by the LOEWE program Ubiquitin Networks (Ub-Net) and the LOEWE Centre for Gene and Cell Therapy Frankfurt, which are both funded by the State of Hessen, Germany (to I.D.). Funding was provided to M.M.E. by Le Fonds de Recherche Santé du Québec and to H.M. by the Alexander von Humboldt foundation as a Humboldt research fellowship for postdoctoral researchers.

Author information

Authors and Affiliations

Authors

Contributions

K.E.Y., H.M., E.F., M.M.E., S.M.V., M.C., D.M. and I.D. designed the conceptual framework of the study and experiments. K.E.Y. and M.M.E. contributed to ENU mutation identification and performed all in vivo and ex vivo experiments involving Cyri mutant and conditional alleles and analysed the data. H.M. designed, performed and analysed all biochemical and cell biology assays. E.F. designed, performed and analysed all mass spectrometry and in vitro Salmonella-infection experiments and performed some biochemical assays. J.A.S. and J.M. performed the analyses of exome sequences. A.A.G. and J.N.M. designed, performed and analysed the experiments with neutrophil mobility assays. D.M. and I.D. supervised the project. All authors wrote the manuscript.

Corresponding authors

Correspondence to Danielle Malo or Ivan Dikic.

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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–13, Supplementary Tables 1–5, Supplementary Notes and Supplementary References.

Reporting Summary

Supplementary File 1

Ubiquitylation sites quantified from three replicate SILAC-diGly proteomics experiments in HeLa Flp-In T-REx GFP–SopE cells, as described in Fig. 7.

Supplementary File 2

Statistics for main and supplementary figures.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yuki, K.E., Marei, H., Fiskin, E. et al. CYRI/FAM49B negatively regulates RAC1-driven cytoskeletal remodelling and protects against bacterial infection. Nat Microbiol 4, 1516–1531 (2019). https://doi.org/10.1038/s41564-019-0484-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0484-8

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