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:

Functional screenings reveal different requirements for host microRNAs in Salmonella and Shigella infection

Abstract

MicroRNAs (miRNAs) are increasingly recognized for their role in infection by bacterial pathogens, although the effect of each individual miRNA remains largely unknown. Here, we used a comparative genome-wide microscopy-based functional screening approach to identify miRNAs controlling infection by two bacterial pathogens—Salmonella enterica serovar Typhimurium and Shigella flexneri. Despite the similarities between these pathogens, we found infections to be controlled by largely non-overlapping subsets of miRNAs, seemingly reflecting different requirements prompted by their distinct intracellular lifestyles. By characterizing a small subset of miRNAs chosen among the strongest inhibitors of Shigella infection, we discovered that miR-3668, miR-4732-5p and miR-6073 exert a selective effect on Shigella infection by impairing bacterial actin-based motility by downregulating N-WASP. Additionally, by identifying let-7i-3p miRNA as a strong inhibitor of Salmonella replication and performing in-depth analysis of its mechanisms of action, we showed that this miRNA specifically inhibits Salmonella infection via modulation of endolysosomal trafficking and the vacuolar environment by targeting the host RGS2 protein. These findings illustrate two paradigms underlying miRNA-mediated regulation of bacterial infection, acting as part of the host response to infection, or as part of bacterial strategies to modulate the host environment and favour pathogenesis.

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: Infection by Salmonella and Shigella are controlled by largely non-overlapping subsets of microRNAs.
Fig. 2: miRNAs regulate different stages of Salmonella and Shigella infection cycles.
Fig. 3: Shigella actin-based motility and intercellular spreading are inhibited by miR-3668, miR-4732-5p and miR-6073.
Fig. 4: Let-7i-3p inhibits Salmonella binding to host cells and bacterial intracellular replication.
Fig. 5: Inhibition of Salmonella replication by let-7i-3p is mediated by RGS2.
Fig. 6: Modulation of endolysosomal trafficking by let-7i-3p is critical for Salmonella infection.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available from the corresponding authors upon reasonable request. The demultiplexed FASTQ files and gene-wise quantifications have been deposited in NCBI’s Gene Expression Omnibus and are accessible through GEO Series accession numbers GSE53281 (mock-treated and Salmonella-infected samples) and GSE69660 (cells transfected with control miRNA or let-7i-3p mimic). Source data for Figs. 3e,i,k, 4f, 5f,g,i and 6h and Extended Data Figs. 3b,d, 5f,h,j and 6b are included in this article and its Supplementary Information files.

Code availability

Custom image analysis workflows implemented in Columbus or CellProfiler image analysis software are available upon reasonable request.

References

  1. Bartel, D. P. Metazoan microRNAs. Cell 173, 20–51 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Jonas, S. & Izaurralde, E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat. Rev. Genet. 16, 421–433 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Friedman, R. C., Farh, K. K., Burge, C. B. & Bartel, D. P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19, 92–105 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Aguilar, C., Mano, M. & Eulalio, A. MicroRNAs at the host–bacteria interface: host defense or bacterial offense. Trends Microbiol. 27, 206–218 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Duval, M., Cossart, P. & Lebreton, A. Mammalian microRNAs and long noncoding RNAs in the host-bacterial pathogen crosstalk. Semin. Cell. Dev. Biol. 65, 11–19 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Eulalio, A. & Mano, M. MicroRNA screening and the quest for biologically relevant targets. J. Biomol. Screen. 20, 1003–1017 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Lemons, D., Maurya, M. R., Subramaniam, S. & Mercola, M. Developing microRNA screening as a functional genomics tool for disease research. Front. Physiol. 4, 223 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Rodrigues Lopes, I., Silva, R. J., Caramelo, I., Eulalio, A. & Mano, M. Shedding light on microRNA function via microscopy-based screening. Methods 152, 55–64 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Huang, Q. et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat. Cell Biol. 10, 202–210 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Eulalio, A. et al. Functional screening identifies miRNAs inducing cardiac regeneration. Nature 492, 376–381 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Judson, R. L., Greve, T. S., Parchem, R. J. & Blelloch, R. MicroRNA-based discovery of barriers to dedifferentiation of fibroblasts to pluripotent stem cells. Nat. Struct. Mol. Biol. 20, 1227–1235 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Santhakumar, D. et al. Combined agonist–antagonist genome-wide functional screening identifies broadly active antiviral microRNAs. Proc. Natl Acad. Sci. USA 107, 13830–13835 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Maudet, C. et al. Functional high-throughput screening identifies the miR-15 microRNA family as cellular restriction factors for Salmonella infection. Nat. Commun. 5, 4718 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Collaborators, G. B. D. D. D. Estimates of global, regional, and national morbidity, mortality, and aetiologies of diarrhoeal diseases: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. Infect. Dis. 17, 909–948 (2017).

    Article  Google Scholar 

  15. Haselbeck, A. H. et al. Current perspectives on invasive nontyphoidal Salmonella disease. Curr. Opin. Infect. Dis. 30, 498–503 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Kotloff, K. L., Riddle, M. S., Platts-Mills, J. A., Pavlinac, P. & Zaidi, A. K. M. Shigellosis. Lancet 391, 801–812 (2018).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mellouk, N. & Enninga, J. Cytosolic access of intracellular bacterial pathogens: the Shigella paradigm. Front. Cell. Infect. Microbiol. 6, 35 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Agaisse, H. Molecular and cellular mechanisms of Shigella flexneri dissemination. Front. Cell. Infect. Microbiol. 6, 29 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Lambrechts, A., Gevaert, K., Cossart, P., Vandekerckhove, J. & Van Troys, M. Listeria comet tails: the actin-based motility machinery at work. Trends Cell Biol. 18, 220–227 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Agarwal, V., Bell, G. W., Nam, J. W. & Bartel, D. P. Predicting effective microRNA target sites in mammalian mRNAs. eLife 4, 05005 (2015).

    Article  Google Scholar 

  22. Egile, C. et al. Activation of the CDC42 effector N-WASP by the Shigella flexneri IcsA protein promotes actin nucleation by Arp2/3 complex and bacterial actin-based motility. J. Cell Biol. 146, 1319–1332 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Suzuki, T. et al. Neural Wiskott-Aldrich syndrome protein (N-WASP) is the specific ligand for Shigella VirG among the WASP family and determines the host cell type allowing actin-based spreading. Cell. Microbiol. 4, 223–233 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Boujemaa-Paterski, R. et al. Listeria protein ActA mimics WASp family proteins: it activates filament barbed end branching by Arp2/3 complex. Biochemistry 40, 11390–11404 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Bierne, H. et al. WASP-related proteins, Abi1 and Ena/VASP are required for Listeria invasion induced by the Met receptor. J. Cell Sci. 118, 1537–1547 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Lara-Tejero, M. & Galan, J. E. Salmonella enterica serovar Typhimurium pathogenicity island 1-encoded type III secretion system translocases mediate intimate attachment to nonphagocytic cells. Infect. Immun. 77, 2635–2642 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Misselwitz, B. et al. Salmonella enterica serovar Typhimurium binds to HeLa cells via Fim-mediated reversible adhesion and irreversible type three secretion system 1-mediated docking. Infect. Immun. 79, 330–341 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Schlumberger, M. C. et al. Real-time imaging of type III secretion: Salmonella SipA injection into host cells. Proc. Natl Acad. Sci. USA 102, 12548–12553 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Menard, R., Sansonetti, P. J. & Parsot, C. Nonpolar mutagenesis of the ipa genes defines IpaB, IpaC, and IpaD as effectors of Shigella flexneri entry into epithelial cells. J. Bacteriol. 175, 5899–5906 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Woodard, G. E., Jardin, I., Berna-Erro, A., Salido, G. M. & Rosado, J. A. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. Int. Rev. Cell Mol. Biol. 317, 97–183 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Sethakorn, N., Yau, D. M. & Dulin, N. O. Non-canonical functions of RGS proteins. Cell. Signal. 22, 1274–1281 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tuli, A. & Sharma, M. How to do business with lysosomes: Salmonella leads the way. Curr. Opin. Microbiol. 47, 1–7 (2018).

    Article  PubMed  CAS  Google Scholar 

  33. Garcia-del Portillo, F. & Finlay, B. B. Targeting of Salmonella typhimurium to vesicles containing lysosomal membrane glycoproteins bypasses compartments with mannose 6-phosphate receptors. J. Cell Biol. 129, 81–97 (1995).

    Article  CAS  PubMed  Google Scholar 

  34. Rathman, M., Barker, L. P. & Falkow, S. The unique trafficking pattern of Salmonella typhimurium-containing phagosomes in murine macrophages is independent of the mechanism of bacterial entry. Infect. Immun. 65, 1475–1485 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Santos, J. C. et al. The COPII complex and lysosomal VAMP7 determine intracellular Salmonella localization and growth. Cell. Microbiol. 17, 1699–1720 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Hang, H. C. et al. Mechanism-based probe for the analysis of cathepsin cysteine proteases in living cells. ACS Chem. Biol. 1, 713–723 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Meresse, S., Steele-Mortimer, O., Finlay, B. B. & Gorvel, J. P. The rab7 GTPase controls the maturation of Salmonella typhimurium-containing vacuoles in HeLa cells. EMBO J. 18, 4394–4403 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Brown, W. J., DeWald, D. B., Emr, S. D., Plutner, H. & Balch, W. E. Role for phosphatidylinositol 3-kinase in the sorting and transport of newly synthesized lysosomal enzymes in mammalian cells. J. Cell Biol. 130, 781–796 (1995).

    Article  CAS  PubMed  Google Scholar 

  39. Davidson, H. W. Wortmannin causes mistargeting of procathepsin D. Evidence for the involvement of a phosphatidylinositol 3-kinase in vesicular transport to lysosomes. J. Cell Biol. 130, 797–805 (1995).

    Article  CAS  PubMed  Google Scholar 

  40. Gaffet, P., Jones, A. T. & Clague, M. J. Inhibition of calcium-independent mannose 6-phosphate receptor incorporation into trans-Golgi network-derived clathrin-coated vesicles by wortmannin. J. Biol. Chem. 272, 24170–24175 (1997).

    Article  CAS  PubMed  Google Scholar 

  41. Ikeda, K. et al. Drug-induced phospholipidosis is caused by blockade of mannose 6-phosphate receptor-mediated targeting of lysosomal enzymes. Biochem. Biophys. Res. Commun. 377, 268–274 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Reaves, B. J., Bright, N. A., Mullock, B. M. & Luzio, J. P. The effect of wortmannin on the localisation of lysosomal type I integral membrane glycoproteins suggests a role for phosphoinositide 3-kinase activity in regulating membrane traffic late in the endocytic pathway. J. Cell Sci. 109, 749–762 (1996).

    Article  CAS  PubMed  Google Scholar 

  43. Brumell, J. H., Tang, P., Zaharik, M. L. & Finlay, B. B. Disruption of the Salmonella-containing vacuole leads to increased replication of Salmonella enterica serovar typhimurium in the cytosol of epithelial cells. Infect. Immun. 70, 3264–3270 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Collado-Romero, M., Arce, C., Ramirez-Boo, M., Carvajal, A. & Garrido, J. J. Quantitative analysis of the immune response upon Salmonella typhimurium infection along the porcine intestinal gut. Vet. Res. 41, 23 (2010).

    Article  PubMed  CAS  Google Scholar 

  45. Denzler, R. et al. Impact of microRNA levels, target-site complementarity, and cooperativity on competing endogenous RNA-regulated gene expression. Mol. Cell 64, 565–579 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hon, L. S. & Zhang, Z. The roles of binding site arrangement and combinatorial targeting in microRNA repression of gene expression. Genome Biol. 8, R166 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Campellone, K. G. & Welch, M. D. A nucleator arms race: cellular control of actin assembly. Nat. Rev. Mol. Cell Biol. 11, 237–251 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sunkavalli, U. et al. Analysis of host microRNA function uncovers a role for miR-29b-2-5p in Shigella capture by filopodia. PLoS Pathog. 13, e1006327 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Hume, P. J., Singh, V., Davidson, A. C. & Koronakis, V. Swiss Army pathogen: the Salmonella entry toolkit. Front. Cell. Infect. Microbiol. 7, 348 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Van Nhieu, G. T. & Romero, S. Common themes in cytoskeletal remodeling by intracellular bacterial effectors. Handb. Exp. Pharmacol. 235, 207–235 (2017).

    Article  PubMed  CAS  Google Scholar 

  51. Sullivan, B. M. et al. RGS4 and RGS2 bind coatomer and inhibit COPI association with Golgi membranes and intracellular transport. Mol. Biol. Cell 11, 3155–3168 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. McGourty, K. et al. Salmonella inhibits retrograde trafficking of mannose-6-phosphate receptors and lysosome function. Science 338, 963–967 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tawk, C. et al. Stress-induced host membrane remodeling protects from infection by non-motile bacterial pathogens. EMBO J. 37, e98529 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Papenfort, K. et al. Specific and pleiotropic patterns of mRNA regulation by ArcZ, a conserved, Hfq-dependent small RNA. Mol. Microbiol. 74, 139–158 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Eulalio, A., Frohlich, K. S., Mano, M., Giacca, M. & Vogel, J. A candidate approach implicates the secreted Salmonella effector protein SpvB in P-body disassembly. PLoS ONE 6, e17296 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Garcia-Feliz, C. et al. Salmonella enterica infections in Spanish swine fattening units. Zoonoses Public Health 54, 294–300 (2007).

    Article  CAS  PubMed  Google Scholar 

  57. Hussain, N. K. et al. Endocytic protein intersectin-l regulates actin assembly via Cdc42 and N-WASP. Nat. Cell Biol. 3, 927–932 (2001).

    Article  CAS  PubMed  Google Scholar 

  58. Sancak, Y. et al. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320, 1496–1501 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. McQuin, C. et al. CellProfiler 3.0: Next-generation image processing for biology. PLoS Biol. 16, e2005970 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Collado-Romero, M. et al. Quantitative proteomics and bioinformatic analysis provide new insight into the dynamic response of porcine intestine to Salmonella Typhimurium. Front. Cell. Infect. Microbiol. 5, 64 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Forstner, K. U., Vogel, J. & Sharma, C. M. READemption-a tool for the computational analysis of deep-sequencing-based transcriptome data. Bioinformatics 30, 3421–3423 (2014).

    Article  PubMed  CAS  Google Scholar 

  62. Otto, C., Stadler, P. F. & Hoffmann, S. Lacking alignments? The next-generation sequencing mapper segemehl revisited. Bioinformatics 30, 1837–1843 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sturn, A., Quackenbush, J. & Trajanoski, Z. Genesis: cluster analysis of microarray data. Bioinformatics 18, 207–208 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

C.A. is a recipient of a fellowship from the Bayerischen Gleichstellungsförderung (BGF) through the SCIENTIA Program; I.R.L. and R.J.S. are recipients of PhD fellowships (PD/BD/146464/2019 and PD/BD/129294/2017) of the Doctoral Programme in Experimental Biology and Biomedicine of the Center for Neuroscience and Cell Biology, University of Coimbra. The authors acknowledge the animal facility staff at the University of Leon, Spain, in particular A. Carvajal from the Department of Animal Health. This work was supported by grants from the Bavarian Ministry of Sciences, Research and the Arts in the framework of the Bavarian Molecular Biosystems Research Network (BioSysNet), DFG project BR 4837/1-1 and the Portuguese Foundation for Science and Technology (FCT Investigator Programme IF/01105/2015).

Author information

Authors and Affiliations

Authors

Contributions

C.A., A.R.C., C.M., M.M. and A.E. designed the experiments; A.R.C., M.M. and A.E. performed the high-throughput screening and validation experiments; C.A., I.R.L., C.M., U.S., R.J.S. and C.L. performed the biochemical and cell biology assays for characterization of the role of miRNAs in infection; M.S. performed the analysis of the RNA-sequencing datasets; S.Z.-L. and J.J.G. performed the in vivo Salmonella infections; M.G. provided reagents and access to equipment; M.M. and A.E. wrote the manuscript, with input from all authors.

Corresponding authors

Correspondence to Miguel Mano or Ana Eulalio.

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 Screenings for miRNAs controlling infection by Salmonella and Shigella.

a. Microscopy images and corresponding image segmentation outlines of HeLa cells treated with control miRNA and infected with Salmonella or Shigella. Cells marked in green in the image segmentation panels correspond to cells with high intracellular bacterial load; cells touching the borders of the images were excluded from the analysis. Scale bar, 100 μm. b and c. Pairwise correlation between the three independent runs of the screenings to identify miRNAs that regulate Salmonella (b) and Shigella (c) infection. Results are based on quantification of the percentage of infected cells following treatment with the library of miRNA mimics and are shown as fold change compared to control miRNA (log2 scale). Spearman rank correlation coefficients are shown in the right bottom corner of graphs. d and e. Comparison of the results obtained in the Salmonella (d) and Shigella (e) screenings for the 110 selected miRNAs, with those obtained for the corresponding time point in the time-course experiments (20 hpi, Salmonella; 6 hpi, Shigella). Results shown are the mean of the three independent runs of each screen or time-course experiments. Colours in panels d and e correspond to the different miRNA categories identified in Figure 2a. Spearman rank correlation coefficients are shown in the right bottom corner of graphs.

Extended Data Fig. 2 Shigella intracellular motility and intercellular spreading are inhibited by miR-3668, miR-4732-5p and miR-6073.

a. Fluorescence microscopy images extracted from the time-lapse microscopy analysis of HeLa cells infected with Shigella wild-type, upon treatment with control miRNA, miR-3668, miR-4732-5p or miR-6073 mimics. Full time-lapse sequence is included as Supplementary Videos 1-4. Scale bar, 100 μm. b and c. Representative images (b) and area of Shigella infection foci (c) in HCT-8 cells transfected with control miRNA, miR-3668, miR-4732-5p or miR-6073 mimics and infected with Shigella, analyzed at 3 hpi. Shigella ΔicsA mutant is shown for comparison. Scale bar, 25 µm. d. Area of Listeria infection foci in HeLa cells transfected with the indicated miRNAs, analyzed at 8 hpi. e. Sequences of mature human miR-3668, miR-4732-5p and miR-6073. The 7-mer seed sequences of the miRNAs are highlighted in red. f. N-WASP levels, determined by western blot, in HeLa cells transfected with control miRNA, miR-3668, miR-4732-5p or miR-6073 mimics, or with control siRNA or N-WASP siRNA. g and h. Representative images (g) and percentage of bacteria forming actin tails (h) in HeLa cells transfected with control siRNA or N-WASP siRNA, analyzed at 1.5 hpi. Staining of F-actin using fluorescently labeled phalloidin allows the visualization of actin tails. Scale bar, 10 µm. i and j. Area of Shigella infection foci in HeLa (i) and HCT-8 (j) cells transfected with control miRNA or N-WASP siRNA, analyzed at 3 hpi. Shigella ΔicsA mutant is shown for comparison. k. Area of Listeria infection foci in HeLa cells transfected with control miRNA or N-WASP siRNA, analyzed at 3 hpi. Shigella infection was performed at MOI 25 (b,g,h), 10 (a,i) or 5 (c,j); Listeria infection was performed at MOI 20. Results are shown as mean ± s.e.m. of n=4 (h) or n=5 (c,f,i-k) biologically independent experiments; microscopy images are representative of n=3 (a), n=4 (g) or n=5 (b) biologically independent experiments; centre values indicate means (c,d,h-j); ***P<0.001 (statistical analyses are detailed in Supplementary Table 4).

Extended Data Fig. 3 Targeting of N-WASP by miR-3668, miR-4732-5p and miR-6073 inhibits Shigella intercellular spreading.

a. Area of Shigella infection foci (in parental HeLa cells, HeLa cells stably expressing a siRNA resistant version of N-WASP fused with EGFP (EGFP-N-WASPr) or wild-type N-WASP fused with EGFP (EGFP-N-WASP), transfected with a control siRNA or a siRNA targeting N-WASP, analyzed at 3 hpi. b. N-WASP expression, determined by western blot, in HeLa cells (parental, EGFP-N-WASPr, EGFP-N-WASP) transfected with a control siRNA or a siRNA targeting N-WASP. c. Area of Shigella infection foci in HeLa cells transfected with control siRNA or 4 independent siRNAs targeting N-WASP, analyzed at 3 hpi. d. N-WASP expression, determined by western blot, in HeLa cells transfected with control siRNA or 4 independent siRNAs targeting N-WASP. e. Sequences of the six active binding sites for miR-3668 (2 sites), miR-4732-5p (1 site) and miR-6073 (3 sites) identified in the 3’UTR of N-WASP. Complementarity of the binding sites with the miRNAs, as predicted by TargetScan, is shown. f. N-WASP levels, determined by western blot, in HeLa cells mock-treated or infected with Shigella, at different times post-infection (0.5, 3, 6 and 8 hpi). g. Expression of mature miR-4732-5p or miR-6073, determined by RT-qPCR, in HeLa cells transfected with control miRNA inhibitor or inhibitors of miR-4732-5p and miR-6073, separately or simultaneously. h. N-WASP levels, determined by western blot, in HeLa cells mock-treated or infected with Shigella, and transfected with control miRNA inhibitor or inhibitors of miR-4732-5p and miR-6073 simultaneously, analyzed at 8 hpi. Shigella infection was performed at MOI 100 (f,h) or 10 (a,c). Results are shown as mean ± s.e.m. of n=3 (g,h) or n=5 (a,c,f) biologically independent experiments; western blots (b,d) are representative of n=3 biologically independent experiments; centre values indicate means (a,d); **P<0.01, ***P<0.001 (statistical analyses are detailed in Supplementary Table 4).

Source Data

Extended Data Fig. 4 Disruption of actin stress fibers reverts the inhibitory effect of let-7i-3p on Salmonella binding to host cells.

a. Sequences of the mature miRNAs derived from the 5’ and 3’ arm of the precursors of the let-7 family. Common seed sequence of the let-7 miRNA family (-5p miRNAs) is highlighted in blue and that of let-7i-3p is highlighted in green. b, c, e and f. Representative images (b,e) and quantification by cfu (c,f) of Salmonella bound to HeLa cells transfected with control miRNA or let-7i-3p mimics, and incubated with Salmonella wild-type or Δ4 mutant strain for 10 min. Cells were pre-treated with cytochalasin D for 1 h (b,c). Salmonella was previously coated with PL (e,f). d, g and h. Representative images (d,g) and quantification by cfu (h) of Shigella bound to HeLa cells transfected with control miRNA or let-7i-3p mimics, and incubated with Shigella wild-type or ΔipaB mutant strains for 25 min. Shigella was previously coated with PL (g,h). i-l. Representative images (i,k) and quantification by cfu (j,l) of Listeria bound to (i,j) or intracellular Listeria (k,l) of HeLa cells transfected with control miRNA or let-7i-3p mimics. Binding was performed for 25 min, and intracellular bacteria analyzed at 1 and 20 hpi. m-p. Expression of mature let-7i-3p (m,n) and RGS2 (o,p), evaluated by RT-qPCR, in HeLa cells infected with Shigella (m,o) or Listeria (n,p). Shigella and Listeria infections were analyzed at 6 or 20 hpi, respectively. Salmonella infection was performed at MOI 50. Shigella infection was performed at MOI 50 (d,g,h) or MOI 100 (m,o). Listeria infection was performed at MOI 25 (k,l,n,p) or 50 (i,j). Scale bars, 50 μm. Results are shown as mean ± s.e.m. of n=5 (c,f,h,l-p) or n=6 (j) biologically independent experiments; microscopy images are representative of n=3 (i,k) or n=5 (b,d,e,g) biologically independent experiments; ***P<0.001 (statistical analyses are detailed in Supplementary Table 4).

Extended Data Fig. 5 Let-7i-3p targets the coding sequence of RGS2.

a. Schematic representation of RGS2 3′UTR, CDS, and CDS fragments and mutant constructs used for miRNA binding site reporter assays. The RGS2 3’UTR was cloned downstream of a Renilla luciferase reporter (psiCHECK2-RGS2 3’UTR) and the RGS2 CDS and CDS fragments were cloned as a C-terminal fusion of green fluorescent protein (EGFP-RGS2 CDS). b-d. Results of the reporter assays using the RGS2 3’UTR (b), RGS2 CDS (c), or RGS2 CDS fragments and mutant (d). Results in cells treated with the let-7i-3p mimic are shown compared to the control miRNA. e. Sequence of the binding site for let-7i-3p in fragment F1 of the N-WASP CDS. Complementarity of the miRNA with the binding site of human RGS2 sequence is shown. f. RGS2 expression, determined by western blot, in HeLa cells transfected with control siRNA or RGS2 siRNA. g. Quantification of intracellular Salmonella by cfu in parental HeLa cells, HeLa cells stably expressing a siRNA resistant version of RGS2 fused with EGFP (EGFP-RGS2r) or wild-type RGS2 fused with EGFP (EGFP-RGS2), transfected with a control siRNA or a siRNA targeting RGS2, analyzed at 20 hpi. h. RGS2 expression, determined by western blot, in HeLa cells (parental, EGFP-RGS2r or EGFP-RGS2), transfected with a control siRNA or a siRNA targeting RGS2. i. Quantification of intracellular Salmonella by cfu in HeLa cells transfected with control siRNA or 4 independent siRNAs targeting RGS2, analyzed at 20 hpi. j. RGS2 expression, determined by western blot, in HeLa cells transfected with control siRNA or 4 independent siRNAs targeting RGS2. Salmonella infection was performed at MOI 25. Results are shown as mean ± s.e.m. of n=3 (b), n=5 (g,i), n=7 (d) or n=10 (c) biologically independent experiments; western blots (f,h,j) are representative of n=3 biologically independent experiments; **P<0.01, ***P<0.001 (statistical analyses are detailed in Supplementary Table 4).

Source Data

Extended Data Fig. 6 Inhibition of let-7i-3p favours Salmonella infection.

a. Expression of mature let-7i-3p, determined by RT-qPCR, in HeLa cells transfected with control miRNA or let-7i-3p inhibitors. b. RGS2 expression, determined by western blot, in HeLa cells transfected with control miRNA or let-7i-3p inhibitors; non-transfected and mock-transfected cells are shown for comparison. c. Representative images of HeLa cells transfected with control miRNA or let-7i-3p inhibitors and infected with Salmonella. Infection was analyzed at 1, 4 and 20 hpi. Salmonella infection was performed at MOI 25. Scale bar, 50 μm. d-f. Representative images of HeLa cells transfected with control miRNA or let-7i-3p inhibitors and stained with Lysotracker (d), for LAMP-1 (e), or with DQ Red BSA (f). Scale bar, 50 μm. Results are shown as mean ± s.e.m. of n=4 (a) biologically independent experiments; western blots and microscopy images are representative of n=3 (b,d-f) or n=5 (c) biologically independent experiments; ***P<0.001 (statistical analyses are detailed in Supplementary Table 4).

Source Data

Supplementary information

Reporting Summary

Supplementary Video 1

Time-lapse microscopy of Shigella infection in HeLa cells transfected with control miRNA. Data are representative of n = 3 biologically independent experiments.

Supplementary Video 2

Time-lapse microscopy of Shigella infection in HeLa cells transfected with miR-3668. Data are representative of n = 3 biologically independent experiments.

Supplementary Video 3

Time-lapse microscopy of Shigella infection in HeLa cells transfected with miR-4732-5p. Data are representative of n = 3 biologically independent experiments.

Supplementary Video 4

Time-lapse microscopy of Shigella infection in HeLa cells transfected with miR-6073. Data are representative of n = 3 biologically independent experiments.

Supplementary Tables

Supplementary Tables 1–4.

Source data

Source Data Fig. 3

Unprocessed Western Blots

Source Data Fig. 4

Unprocessed Western Blots

Source Data Fig. 5

Unprocessed Western Blots

Source Data Fig. 6

Unprocessed Western Blots

Source Data Extended Data Fig. 3

Unprocessed Western Blots

Source Data Extended Data Fig. 5

Unprocessed Western Blots

Source Data Extended Data Fig. 6

Unprocessed Western Blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Aguilar, C., Cruz, A.R., Rodrigues Lopes, I. et al. Functional screenings reveal different requirements for host microRNAs in Salmonella and Shigella infection. Nat Microbiol 5, 192–205 (2020). https://doi.org/10.1038/s41564-019-0614-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0614-3

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