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:

Kinase-independent synthesis of 3-phosphorylated phosphoinositides by a phosphotransferase

Abstract

Despite their low abundance, phosphoinositides play a central role in membrane traffic and signalling. PtdIns(3,4,5)P3 and PtdIns(3,4)P2 are uniquely important, as they promote cell growth, survival and migration. Pathogenic organisms have developed means to subvert phosphoinositide metabolism to promote successful infection and their survival in host organisms. We demonstrate that PtdIns(3,4)P2 is a major product generated in host cells by the effectors of the enteropathogenic bacteria Salmonella and Shigella. Pharmacological, gene silencing and heterologous expression experiments revealed that, remarkably, the biosynthesis of PtdIns(3,4)P2 occurs independently of phosphoinositide 3-kinases. Instead, we found that the Salmonella effector SopB, heretofore believed to be a phosphatase, generates PtdIns(3,4)P2 de novo via a phosphotransferase/phosphoisomerase mechanism. Recombinant SopB is capable of generating PtdIns(3,4,5)P3 and PtdIns(3,4)P2 from PtdIns(4,5)P2 in a cell-free system. Through a remarkable instance of convergent evolution, bacterial effectors acquired the ability to synthesize 3-phosphorylated phosphoinositides by an ATP- and kinase-independent mechanism, thereby subverting host signalling to gain entry and even provoke oncogenic transformation.

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: Rapid and sustained PtdIns(3,4)P2 synthesis during Salmonella entry and maturation.
Fig. 2: SopB is necessary and sufficient for PtdIns(3,4)P2 biosynthesis in mammalian cells.
Fig. 3: Class I PI3Ks and class II PI3K-C2α are not required for PtdIns(3,4)P2 synthesis during Salmonella entry.
Fig. 4: PtdIns(3,4)P2 generation in S. cerevisiae that are devoid of PI3K activity.
Fig. 5: PtdIns(4,5)P2 levels correlate inversely with SopB-mediated PtdIns(3,4)P2 formation.
Fig. 6: SopB requires a PLC-sensitive inositide to generate PtdIns(3,4)P2.
Fig. 7: In vitro reconstitution reveals SopB phosphotransferase activity.
Fig. 8: Time-resolved HPLC–MS analysis of SopB phosphotransferase-phosphatase activities.

Similar content being viewed by others

Data availability

Data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

Custom macros used in this study have been deposited on GitHub and are available for download at the manuscript repository (https://github.com/walpoleg/Walpole-et-al-Nature-Cell-Biology-2022).

References

  1. Balla, T. Phosphoinositides: tiny lipids with giant impact on cell regulation. Physiol. Rev. 93, 1019–1137 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hakim, S., Bertucci, M. C., Conduit, S. E., Vuong, D. L. & Mitchell, C. A. Inositol polyphosphate phosphatases in human disease. Curr. Top. Microbiol. Immunol. 362, 247–314 (2012).

    CAS  PubMed  Google Scholar 

  3. Sasaki, T. et al. Mammalian phosphoinositide kinases and phosphatases. Prog. Lipid Res. 48, 307–343 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Stephens, L. R., Jackson, T. R. & Hawkins, P. T. Agonist-stimulated synthesis of phosphatidylinositol(3,4,5)-trisphosphate: a new intracellular signalling system? Biochim. Biophys. Acta 1179, 27–75 (1993).

    Article  CAS  PubMed  Google Scholar 

  5. Bilanges, B., Posor, Y. & Vanhaesebroeck, B. PI3K isoforms in cell signalling and vesicle trafficking. Nat. Rev. Mol. Cell Biol. 20, 515–534 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Hawkins, P. T. & Stephens, L. R. Emerging evidence of signalling roles for PI(3,4)P2 in Class I and II PI3K-regulated pathways. Biochem. Soc. Trans. 44, 307–314 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Fruman, D. A. & Rommel, C. PI3K and cancer: lessons, challenges and opportunities. Nat. Rev. Drug Discov. 13, 140–156 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lien, E. C., Dibble, C. C. & Toker, A. PI3K signaling in cancer: beyond AKT. Curr. Opin. Cell Biol. 45, 62–71 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Walpole, G. F. W. & Grinstein, S. Endocytosis and the internalization of pathogenic organisms: focus on phosphoinositides. F1000Research 9, 368 (2020).

    Article  CAS  Google Scholar 

  10. Pizarro-Cerdá, J., Kühbacher, A. & Cossart, P. Phosphoinositides and host–pathogen interactions. Biochim. Biophys. Acta 1851, 911–918 (2015).

    Article  PubMed  CAS  Google Scholar 

  11. Steele-Mortimer, O. et al. Activation of Akt/protein kinase B in epithelial cells by the Salmonella typhimurium effector sigD. J. Biol. Chem. 275, 37718–37724 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Marcus, S. L., Wenk, M. R., Steele-Mortimer, O. & Finlay, B. B. A synaptojanin-homologous region of Salmonella typhimurium SigD is essential for inositol phosphatase activity and Akt activation. FEBS Lett. 494, 201–207 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Niebuhr, K. et al. Conversion of PtdIns(4,5)P2 into PtdIns(5)P by the S. flexneri effector IpgD reorganizes host cell morphology. EMBO J. 21, 5069–5078 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Pendaries, C. et al. PtdIns5P activates the host cell PI3-kinase/Akt pathway during Shigella flexneri infection. EMBO J. 25, 1024–1034 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ebner, M., Lučić, I., Leonard, T. A. & Yudushkin, I. PI(3,4,5)P3 engagement restricts Akt activity to cellular membranes. Mol. Cell 65, 416–431 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Manna, D., Albanese, A., Park, W. S. & Cho, W. Mechanistic basis of differential cellular responses of phosphatidylinositol 3,4-bisphosphate- and phosphatidylinositol 3,4,5-trisphosphate-binding pleckstrin homology domains. J. Biol. Chem. 282, 32093–32105 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Goulden, B. D. et al. A high-avidity biosensor reveals plasma membrane PI(3,4)P2 is predominantly a class I PI3K signaling product. J. Cell Biol. 218, 1066–1079 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Liu, S.-L. et al. Quantitative lipid imaging reveals a new signaling function of phosphatidylinositol-3,4-bisphophate: isoform- and site-specific activation of Akt. Mol. Cell 71, 1092–1104 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Dowler, S. et al. Identification of pleckstrin-homology-domain-containing proteins with novel phosphoinositide-binding specificities. Biochem. J. 351, 19–31 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Thomas, C. C., Dowler, S., Deak, M., Alessi, D. R. & van Aalten, D. M. Crystal structure of the phosphatidylinositol 3,4-bisphosphate-binding pleckstrin homology (PH) domain of tandem PH-domain-containing protein 1 (TAPP1): molecular basis of lipid specificity. Biochem. J. 358, 287–294 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Branchu, P., Bawn, M. & Kingsley, R. A. Genome variation and molecular epidemiology of Salmonella enterica serovar Typhimurium pathovariants. Infect. Immun. 86, e00079-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Posor, Y. et al. Spatiotemporal control of endocytosis by phosphatidylinositol-3,4-bisphosphate. Nature 499, 233–237 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Gewinner, C. et al. Evidence that inositol polyphosphate 4-phosphatase type II is a tumor suppressor that inhibits PI3K signaling. Cancer Cell 16, 115–125 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Norris, F. A. & Majerus, P. W. Hydrolysis of phosphatidylinositol 3,4-bisphosphate by inositol polyphosphate 4-phosphatase isolated by affinity elution chromatography. J. Biol. Chem. 269, 8716–8720 (1994).

    Article  CAS  PubMed  Google Scholar 

  26. Klarlund, J. K., Tsiaras, W., Holik, J. J., Chawla, A. & Czech, M. P. Distinct polyphosphoinositide binding selectivities for pleckstrin homology domains of GRP1-like proteins based on diglycine versus triglycine motifs. J. Biol. Chem. 275, 32816–32821 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Cronin, T. C., DiNitto, J. P., Czech, M. P. & Lambright, D. G. Structural determinants of phosphoinositide selectivity in splice variants of Grp1 family PH domains. EMBO J. 23, 3711–3720 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Venkateswarlu, K., Oatey, P. B., Tavaré, J. M. & Cullen, P. J. Insulin-dependent translocation of ARNO to the plasma membrane of adipocytes requires phosphatidylinositol 3-kinase. Curr. Biol. 8, 463–466 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Galán, J. E., Lara-Tejero, M., Marlovits, T. C. & Wagner, S. Bacterial type III secretion systems: specialized nanomachines for protein delivery into target cells. Annu. Rev. Microbiol. 68, 415–438 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Rodríguez-Escudero, I., Ferrer, N. L., Rotger, R., Cid, V. J. & Molina, M. Interaction of the Salmonella Typhimurium effector protein SopB with host cell Cdc42 is involved in intracellular replication. Mol. Microbiol. 80, 1220–1240 (2011).

    Article  PubMed  CAS  Google Scholar 

  31. Marcus, S. L., Knodler, L. A. & Finlay, B. B. Salmonella enterica serovar Typhimurium effector SigD/SopB is membrane-associated and ubiquitinated inside host cells. Cell. Microbiol. 4, 435–446 (2002).

    Article  CAS  PubMed  Google Scholar 

  32. Galyov, E. E. et al. A secreted effector protein of Salmonella dublin is translocated into eukaryotic cells and mediates inflammation and fluid secretion in infected ileal mucosa. Mol. Microbiol. 25, 903–912 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Anderson Norris, F., Wilson, M. P., Wallis, T. S., Galyov, E. E. & Majerus, P. W. SopB, a protein required for virulence of Salmonella dublin, is an inositol phosphate phosphatase. Proc. Natl Acad. Sci. USA 95, 14057–14059 (1998).

    Article  PubMed Central  Google Scholar 

  34. Zhang, S. et al. The Salmonella enterica serotype typhimurium effector proteins SipA, SopA, SopB, SopD, and SopE2 act in concert to induce diarrhea in calves. Infect. Immun. 70, 3843–3855 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tahoun, A. et al. Salmonella transforms follicle-associated epithelial cells into M cells to promote intestinal invasion. Cell Host Microbe 12, 645–656 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Kum, W. W. S., Lo, B. C., Yu, H. B. & Finlay, B. B. Protective role of Akt2 in Salmonella enterica serovar Typhimurium-induced gastroenterocolitis. Infect. Immun. 79, 2554–2566 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rogers, L. D., Brown, N. F., Fang, Y., Pelech, S. & Foster, L. J. Phosphoproteomic analysis of Salmonella-infected cells identifies key kinase regulators and SopB-dependent host phosphorylation events. Sci. Signal. 4, 1–14 (2011).

    Article  CAS  Google Scholar 

  38. Feng, Y., Wente, S. R. & Majerus, P. W. Overexpression of the inositol phosphatase SopB in human 293 cells stimulates cellular chloride influx and inhibits nuclear mRNA export. Proc. Natl Acad. Sci. USA 98, 875–879 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cooper, K. G. et al. Activation of Akt by the bacterial inositol phosphatase, SopB, is wortmannin insensitive. PLoS ONE 6, e22260 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhou, D., Chen, L. M., Hernandez, L., Shears, S. B. & Galan, J. E. A Salmonella inositol polyphosphatase acts in conjunction with other bacterial effectors to promote host cell actin cytoskeleton rearrangements and bacterial internalization. Mol. Microbiol. 39, 248–259 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Raffatellu, M. et al. SipA, SopA, SopB, SopD, and SopE2 contribute to Salmonella enterica serotype Typhimurium invasion of epithelial cells. Infect. Immun. 73, 146–154 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rahdar, M. et al. A phosphorylation-dependent intramolecular interaction regulates the membrane association and activity of the tumor suppressor PTEN. Proc. Natl Acad. Sci. USA 106, 480–485 (2009).

    Article  CAS  PubMed  Google Scholar 

  43. Luo, J. et al. Genetically encoded optochemical probes for simultaneous fluorescence reporting and light activation of protein function with two-photon excitation. J. Am. Chem. Soc. 136, 15551–15558 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Courtney, T. M. & Deiters, A. Optical control of protein phosphatase function. Nat. Commun. 10, 4384 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Arcaro, A. & Wymann, M. P. Wortmannin is a potent phosphatidylinositol 3-kinase inhibitor: the role of phosphatidylinositol 3,4,5-trisphosphate in neutrophil responses. Biochem. J. 296, 297–301 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Knight, Z. A. et al. A pharmacological map of the PI3-K family defines a role for p110α in insulin signaling. Cell 125, 733–747 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Folkes, A. J. et al. The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer. J. Med. Chem. 51, 5522–5532 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Domin, J. et al. Cloning of a human phosphoinositide 3-kinase with a C2 domain that displays reduced sensitivity to the inhibitor wortmannin. Biochem. J. 326, 139–147 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Virbasius, J. V., Guilherme, A. & Czech, M. P. Mouse p170 is a novel phosphatidylinositol 3-kinase containing a C2 domain. J. Biol. Chem. 271, 13304–13307 (1996).

    Article  CAS  PubMed  Google Scholar 

  50. Schu, P. V. et al. Phosphatidylinositol 3-kinase encoded by yeast VPS34 gene essential for protein sorting. Science 260, 88–91 (1993).

    Article  CAS  PubMed  Google Scholar 

  51. Vanhaesebroeck, B., Guillermet-Guibert, J., Graupera, M. & Bilanges, B. The emerging mechanisms of isoform-specific PI3K signalling. Nat. Rev. Mol. Cell Biol. 11, 329–341 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Alemán, A. et al. The amino-terminal non-catalytic region of Salmonella typhimurium SigD affects actin organization in yeast and mammalian cells. Cell. Microbiol. 7, 1432–1446 (2005).

    Article  PubMed  CAS  Google Scholar 

  53. Herman, P. K. & Emr, S. D. Characterization of VPS34, a gene required for vacuolar protein sorting and vacuole segregation in Saccharomyces cerevisiae. Mol. Cell. Biol. 10, 6742–6754 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Stack, J. H., DeWald, D. B., Takegawa, K. & Emr, S. D. Vesicle-mediated protein transport: regulatory interactions between the Vps15 protein kinase and the Vps34 PtdIns 3-kinase essential for protein sorting to the vacuole in yeast. J. Cell Biol. 129, 321–334 (1995).

    Article  CAS  PubMed  Google Scholar 

  55. Terebiznik, M. R. et al. Elimination of host cell PtdIns(4,5)P2 by bacterial SigD promotes membrane fission during invasion by Salmonella. Nat. Cell Biol. 4, 766–773 (2002).

    Article  CAS  PubMed  Google Scholar 

  56. Hammond, G. R. V., Machner, M. P. & Balla, T. A novel probe for phosphatidylinositol 4-phosphate reveals multiple pools beyond the Golgi. J. Cell Biol. 205, 113–126 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Várnai, P. & Balla, T. Visualization of phosphoinositides that bind pleckstrin homology domains: calcium- and agonist-induced dynamic changes and relationship to myo-[3H]inositol-labeled phosphoinositide pools. J. Cell Biol. 143, 501–510 (1998).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Stauffer, T. P., Ahn, S. & Meyer, T. Receptor-induced transient reduction in plasma membrane PtdIns(4,5)P2 concentration monitored in living cells. Curr. Biol. 8, 343–346 (1998).

    Article  CAS  PubMed  Google Scholar 

  59. Won, D. H. et al. PI(3,4,5)P3 and PI(4,5)P2 lipids target proteins with polybasic clusters to the plasma membrane. Science 314, 1458–1461 (2006).

    Article  CAS  Google Scholar 

  60. Varnai, P., Thyagarajan, B., Rohacs, T. & Balla, T. Rapidly inducible changes in phosphatidylinositol 4,5-bisphosphate levels influence multiple regulatory functions of the lipid in intact living cells. J. Cell Biol. 175, 377–382 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Serunian, L. A. et al. Polyphosphoinositides produced by phosphatidylinositol 3-kinase are poor substrates for phospholipases C from rat liver and bovine brain. J. Biol. Chem. 264, 17809–17815 (1989).

    Article  CAS  PubMed  Google Scholar 

  62. Willars, G. B., Nahorski, S. R. & Challiss, R. A. J. Differential regulation of muscarinic acetylcholine receptor-sensitive polyphosphoinositide pools and consequences for signaling in human neuroblastoma cells. J. Biol. Chem. 273, 5037–5046 (1998).

    Article  CAS  PubMed  Google Scholar 

  63. Gregory, J. D. The stability of N-ethylmaleimide and its reaction with sulfhydryl groups. J. Am. Chem. Soc. 77, 3922–3923 (1955).

    Article  CAS  Google Scholar 

  64. Malek, M. et al. PTEN regulates PI(3,4)P2 signaling downstream of Class I PI3K. Mol. Cell 68, 566–580 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Clark, J. et al. Quantification of PtdInsP3 molecular species in cells and tissues by mass spectrometry. Nat. Methods 8, 267–272 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Schroeder, G. N. & Hilbi, H. Molecular pathogenesis of Shigella spp.: controlling host cell signaling, invasion, and death by type III secretion. Clin. Microbiol. Rev. 21, 134–156 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Finn, C. E., Chong, A., Cooper, K. G., Starr, T. & Steele-Mortimer, O. A second wave of Salmonella T3SS1 activity prolongs the lifespan of infected epithelial cells. PLoS Pathog. 13, e1006354 (2017).

  68. Knodler, L. A., Finlay, B. B. & Steele-Mortimer, O. The Salmonella effector protein SopB protects epithelial cells from apoptosis by sustained activation of Akt. J. Biol. Chem. 280, 9058–9064 (2005).

    Article  CAS  PubMed  Google Scholar 

  69. Hu, G. Q. et al. Salmonella outer protein B suppresses colitis development via protecting cell from necroptosis. Front. Cell. Infect. Microbiol. 9, 87 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hu, G. Q. et al. Cirtical role for Salmonella effector SopB in regulating inflammasome activation. Mol. Immunol. 90, 280–286 (2017).

    Article  CAS  PubMed  Google Scholar 

  71. Zhang, K. et al. Minimal SPI1-T3SS effector requirement for Salmonella enterocyte invasion and intracellular proliferation in vivo. PLoS Pathog. 14, e1006925 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Bruno, V. M. et al. Salmonella typhimurium type III secretion effectors stimulate innate immune responses in cultured epithelial cells. PLoS Pathog. 5, 1000538 (2009).

    Article  CAS  Google Scholar 

  73. Scanu, T. et al. Salmonella manipulation of host signaling pathways provokes cellular transformation associated with gallbladder carcinoma. Cell Host Microbe 17, 763–774 (2015).

    Article  CAS  PubMed  Google Scholar 

  74. Roppenser, B. et al. Multiple host kinases contribute to Akt activation during Salmonella infection. PLoS ONE 8, e71015 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Hsu, F. & Mao, Y. The structure of phosphoinositide phosphatases: insights into substrate specificity and catalysis. Biochim. Biophys. Acta 1851, 698–710 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Mallo, G. V. et al. SopB promotes phosphatidylinositol 3-phosphate formation on Salmonella vacuoles by recruiting Rab5 and Vps34. J. Cell Biol. 182, 741–752 (2008).

  77. Mason, D. et al. Alteration of epithelial structure and function associated with PtdIns(4,5)P2 degradation by a bacterial phosphatase. J. Gen. Physiol. 129, 267–283 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Baek, M. et al. Accurate prediction of protein structures and interactions using a three-track neural network. Science 373, 871–876 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhu, L., Jorgensen, J. R., Li, M., Chuang, Y. S. & Emr, S. D. ESCRTS function directly on the lysosome membrane to downregulate ubiquitinated lysosomal membrane proteins. eLife 6, 1–20 (2017).

    Article  Google Scholar 

  80. Wennström, S. & Downward, J. Role of phosphoinositide 3-kinase in activation of Ras and mitogen-activated protein kinase by epidermal growth factor. Mol. Cell. Biol. 19, 4279–4288 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Leibiger, B. et al. Insulin-feedback via PI3K-C2α activated PKBα/Akt1 is required for glucose-stimulated insulin secretion. FASEB J. 24, 1824–1837 (2010).

    Article  CAS  PubMed  Google Scholar 

  82. Levin, R. et al. Multiphasic dynamics of phosphatidylinositol 4-phosphate during phagocytosis. Mol. Biol. Cell 28, 128–140 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Inoue, T., Heo, W. D., Grimley, J. S., Wandless, T. J. & Meyer, T. An inducible translocation strategy to rapidly activate and inhibit small GTPase signaling pathways. Nat. Methods 2, 415–418 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ran, F. A. et al. Genome engineering using the CRISPR–Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hoiseth, S. K. & Stocker, B. A. Aromatic-dependent Salmonella typhimurium are non-virulent and effective as live vaccines. Nature 291, 238–239 (1981).

    Article  CAS  PubMed  Google Scholar 

  86. Stender, S. et al. Identification of SopE2 from Salmonella typhimurium, a conserved guanine nucleotide exchange factor for Cdc42 of the host cell. Mol. Microbiol. 36, 1206–1221 (2000).

    Article  CAS  PubMed  Google Scholar 

  87. Knodler, L. A. et al. Salmonella effectors within a single pathogenicity island are differentially expressed and translocated by separate type III secretion systems. Mol. Microbiol. 43, 1089–1103 (2002).

    Article  CAS  PubMed  Google Scholar 

  88. Valdivia, R. H., Hromockyj, A. E., Monack, D., Ramakrishnan, L. & Falkow, S. Applications for green fluorescent protein (GFP) in the study of host–pathogen interactions. Gene 173, 47–52 (1996).

    Article  CAS  PubMed  Google Scholar 

  89. Steele-Mortimer, O., Meresse, S., Gorvel, J. P., Toh, B. H. & Finlay, B. B. Biogenesis of Salmonella typhimurium-containing vacuoles in epithelial cells involves interactions with the early endocytic pathway. Cell. Microbiol. 1, 33–49 (1999).

    Article  CAS  PubMed  Google Scholar 

  90. Robinson, J. S., Klionsky, D. J., Banta, L. M. & Emr, S. D. Protein sorting in Saccharomyces cerevisiae: isolation of mutants defective in the delivery and processing of multiple vacuolar hydrolases. Mol. Cell. Biol. 8, 4936–4948 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Horger, K. S., Estes, D. J., Capone, R. & Mayer, M. Films of agarose enable rapid formation of giant liposomes in solutions of physiologic ionic strength. J. Am. Chem. Soc. 131, 1810–1819 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. MacDonald, R. C. et al. Small-volume extrusion apparatus for preparation of large, unilamellar vesicles. Biochim. Biophys. Acta 1061, 297–303 (1991).

    Article  CAS  PubMed  Google Scholar 

  93. Smal, I., Loog, M., Niessen, W. & Meijering, E. Quantitative comparison of spot detection methods in fluorescence microscopy. IEEE Trans. Med. Imaging 29, 282–301 (2010).

    Article  PubMed  Google Scholar 

  94. Olivo-Marin, J.-C. Extraction of spots in biological images using multiscale products. Pattern Recognit. 35, 1989–1996 (2002).

    Article  Google Scholar 

  95. Lord, S. J., Velle, K. B., Mullins, R. D. & Fritz-Laylin, L. K. SuperPlots: Communicating reproducibility and variability in cell biology. J. Cell Biol. 219, e202001064 (2020).

Download references

Acknowledgements

We thank S. Emr (Department of Molecular Biology and Genetics, Cornell University) for sharing plasmids and yeast strains for this study. We thank K. Lau and P. Paroutis (The Imaging Facility, The Hospital for Sick Children) for technical training and discussions of analyses. Models (Figs. 1c,2d,3a,3h,8i and Extended Data Figs. 3a,3b,4b,8e) were created with BioRender.com. G.F.W.W. is supported by a Vanier Canada Graduate Scholarship from the Canadian Institutes of Health Research (CIHR) and an MD/PhD Studentship from the University of Toronto. J.H.B. is supported by CIHR grant no. FDN-154329. A.D. is funded by grant no. R01GM132565 from the National Institutes of Health. G.R.V.H. is funded by grant no. 1R35GM119412-01 from the National Institutes of Health. S.G. is supported by CIHR grant no. FDN-143202, and G.D.F. is supported by CIHR project grant no. PJT165968 and the Natural Sciences and Engineering Research Council (NSERC) of Canada.

Author information

Authors and Affiliations

Authors

Contributions

G.F.W.W., S.G., G.R.V.H. and G.D.F. conceived experiments and developed methods. G.F.W.W. conducted experiments and analysed the resulting data. J.P. conducted photoactivation assays and analysed the resulting data with G.R.V.H. K.E.A. and J.C. performed HPLC–MS measurements and analysed the resulting data with L.R.S. and P.T.H. D.B.-K. performed invasion efficiency assays. N.C., Y.M.A., F.M.-R., Z.L., H.Z., J.H.B. and A.D. made and/or provided critical reagents. G.F.W.W. wrote the original draft of the manuscript. All authors reviewed and edited the manuscript.

Corresponding authors

Correspondence to Sergio Grinstein or Gregory D. Fairn.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Rapid and sustained PtdIns(3,4)P2 synthesis during Salmonella entry and maturation.

(a) Model of PtdIns(3,4)P2 biosensors based on single, double-, or triple-tandem carboxy-terminal PH domains from TAPP1 (PLEKHA1). NES, nuclear export signal. Right, gel electrophoresis of PCR amplicons generated by primers that house the open reading frames. (b) Cells expressing cPHx1, cPHx2, or cPHx3 were infected for 10 min with wild-type Salmonella prior to staining the PM with CellMask. Representative maximum intensity projections (main) and a corresponding confocal section of the invasion ruffle (bottom panels) are presented. (c) Confocal imaging of cells (three examples in I, II, and III) expressing cPHx3 during invasion by wild-type Salmonella. Bottom vertical panels are expanded from the white box region and correspond to the minute-by-minute time series. cPHx3 is also presented in a grey inverted lookup table (RGB intensity 0=white, 255=black). (d) As in (c), three examples (I, II, and III) of cells expressing the PtdIns(3,4,5)P3 sensor aPHx2 during invasion by wild-type Salmonella. (e) Cells serum-starved for 3 h were infected by Salmonella. Extracellular bacteria were removed, and cells returned to serum-free medium containing gentamycin. PM cPHx3 intensities were quantified in the following number of cells: 59 (control), 60 (WT, 30 min), 42 (∆sopB, 30 min), 58 (WT, 60 min), 53 (∆sopB, 60 min), 75 (WT, 120 min), 48 (∆sopB, 120 min), 66 (WT, 240 min), and 36 (∆sopB, 240 min) across n=3 independent experiments. Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (background). Data from Fig. 1c are presented with ∆sopB infections. ****P < 0.0001; **P = 0.0054 (UI-vs-WT60), **P = 0.0044 (WT120-vs-∆sopB120); ns, not significant. (f) Cells serum-starved for 3 hours, were exposed to Salmonella (wild-type or ∆sopB) for 10 min. Extracellular bacteria were removed, and cells were returned to serum-free medium with gentamycin. Lysates were collected at the indicated time points and analysed on parallel membranes for pAKT (S473) or pAKT (T308) prior to stripping and re-probing for total AKT or GAPDH (loading control). Representative immunoblots are presented from n=2 independent experiments. UI, uninfected. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 2 PtdIns(3,4,5)P3 analysis during Salmonella invasion or during optogenetic activation of SopB.

(a) Model of the PtdIns(3,4,5)P3 biosensor NES-EGFP-aPHx2 derived from tandem ARNO PH domains (2G splice variant, I303E mutation). NES, nuclear export signal. (b) Cells were exposed to invasive RFP-expressing wild-type or isogenic ∆sopB Salmonella and the PM was stained with CellMask prior to imaging. Maximum intensity projections (main) and single confocal sections of the invasion ruffle are presented at right for each. (c) Quantification from (b). Normalized intensity of aPHx2 in the PM of invasion sites from n=3 independent experiments analysing 71 (WT) and 63 (∆sopB) invasion sites. *P = 0.0294. (d) Normalized intensity of aPHx2 in the PM during optogenetic activation of SopBWT-464TAG or SopBC460S-464TAG. Filtered baseline-corrected time-lapse data are mean ± s.e.m. of individual cell measurements from 3 independent experiments quantifying n=29 cells (WT) and n=13 cells (C460S). (e) AUC calculations of aPHx2 intensities from (d). Data are median, box (25th-75th) and whisker (10th-90th) percentiles of n=29 cells (WT) and n=13 cells (C460S) from 3 independent experiments. P = 0.4415, ns. (f,g) SopB-mediated synthesis of PtdIns(3,4)P2 is not accompanied by a robust PtdIns(3,4,5)P3 response. Representative confocal sections of aPHx2 and cPHx3 localization during optogenetic activation of WT or C460S SopB. Left, corresponding linear RGB intensity scales are presented. The indicated times are prior to (t - 30 s) or after illumination with 405 nm light to photolyze hydroxycoumarin lysine in SopB. See corresponding quantification of (f) presented in Fig. 2g. Source numerical data are available in source data.

Source data

Extended Data Fig. 3 Sequence determinants of SopB-mediated PtdIns(3,4)P2 generation.

(a) Bacterially injected SopB localizes to the vacuolar membrane and PM invaginations. HeLa cells were exposed to ∆sopB (control) or ∆sopB + SopB-c-myc Salmonella for 10 min prior to labelling the PM (WGA) and immunostaining against c-myc and Salmonella. Inset panels are expanded from the hashed box region. Representative maximum intensity projections are from n=3 independent experiments. (b) Anti-myc fluorescence is not contaminated by non-secreted SopB or anti-Salmonella immunostaining. ∆sopB + SopB-c-myc Salmonella were centrifuged onto poly-L-lysine coated coverslips. Bacteria were processed as in (a) and imaged with equivalent acquisition settings. The representative maximum intensity projection is from n=3 independent experiments. (c) Heterologous SopB coalesces within puncta that abut cortical membranes. Full-length SopB constructs were co-transfected with cPHx3 and imaged live. Amino-terminally tagged SopB was enriched along cytosolic reticular structures; amino- and carboxy-terminal EGFP fusions also enriched on the basal footprint of cells. Representative confocal sections are presented for each construct, from more than three similar experiments. CellMask served as a PM marker. (d) SopB requires amino acids 68-172 and 520-554 for PtdIns(3,4)P2 generation. The indicated SopB plasmids (top panels) were co-transfected with cPHx3 (bottom panels) and imaged live in HeLa cells. Representative confocal sections are presented. Note that membrane-targeting of SopB is disrupted following the deletion of amino acids 68-172 while PM-targeting is preserved following the deletion of amino acids 520-554. (e) CRISPR–Cas9-mediated deletion of Cdc42. Total lysates from control sgRNA- or Cdc42-specific sgRNA-treated Henle 407 cells were immunoblotted against Cdc42. Alpha tubulin served as a loading control. (f) Cdc42 regulates SopB targeting but is not strictly required for PtdIns(3,4)P2 generation. Representative maximum intensity projections are presented of SopB (1-561)-EGFP localization in parental wild-type Henle (top) or Cdc42 KO Henle cells (bottom). Inset images depict localization of cPHx3 (confocal section). cPHx3 was markedly enriched in the PM with a concomitant decrease in cytosolic fluorescence in (mean ± s.e.m.) 94.1 ± 2.92% of parental WT and 88.5 ± 7.28% of Cdc42 KO cells across n=3 independent experiments. (g) Quantification of SopB-EGFP puncta per cell from n=3 independent experiments analysing the following number of Henle 407 cells: 91 (WT) and 59 (Cdc42 KO). Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). **P = 0.0014. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 4 3-phosphorylated phosphoinositides promote bacterial invasion of host cells.

(a) Cells were exposed to wild-type or ∆sopB Salmonella for 10 min and extracellular bacteria were removed by extensive washing before returning to growth medium for an additional 20 min. Extracellular and intracellular bacteria were differentially stained. Data are mean ± s.e.m. from n=3 independent experiments quantifying ≥300 infected cells per strain. *P = 0.0333. (b,c) PtdIns(3,4)P2 and/or PtdIns(3,4,5)P2 promote bacterial invasion. (b) Model of PTEN-catalysed reactions: membrane-associated PTEN dephosphorylates both PtdIns(3,4,5)P3 and PtdIns(3,4)P2 at the 3-position of the inositol ring. (c) Cells transfected with EGFP (control) or the A4 mutant of PTEN were left untreated or pre-treated with LY294002 (10 μM, 30 min) prior to invasion. Twofold excess of ∆sopE/sopE2 bacteria was used to obtain sufficient infected cells. Data are mean ± s.e.m. (normalized to respective control) from n=3 independent experiments quantifying ≥300 infected cells per strain. WT: **P = 0.0022, ***P = 0.0007; ∆sopE/E2: *P = 0.0125, **P = 0.0039. (d) Cells were co-transfected with cPHx3 and PM-targeted catalytic subunit of class IA PI3K (p110α-CAAX). Images were acquired immediately before (left) or 3 min after addition of DMSO (vehicle, right). (e) HeLa cells expressing vector control or p110α-CAAX were treated for 20 min with DMSO, wortmannin (100 nM), PI-103 (500 nM), or GDC-0941 (500 nM) prior to collection of cell lysates and immunoblotting using phospho-AKT (S473) and pan AKT antibodies. The immunoblot presented is representative of n=3 independent experiments. Corresponding quantification, Fig. 3c. (f) Confirmation of anti-PI3K-C2α polyclonal antibody labelling of heterologously-expressed PI3K-C2α. HeLa cells transfected with PM-targeted myc-PI3K-C2α were co-stained for the myc epitope tag and PI3K-C2α. Panels right depict enlargement of hashed box regions (I, II), where the non-transfected cells (I) were overexposed to visualize endogenous staining. (g) Class II PI3K-C2α is not enriched at the site of Salmonella invasion. Confocal sections of uninfected and wild-type Salmonella infected HeLa cells stained for endogenous PI3K-C2α. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 5 Lysine residues within the carboxyl-terminus of SopB support optimal phosphotransferase activity.

(a) Alignment of SopB amino acids 504-554 with other PtdIns(4,5)P2 and PtdIns(3,4,5)P3, 5-phosphatases. Basic residues in SopB are highlighted red along with conserved basic residues in the other phosphatases; additional conserved residues are highlighted purple, and regions of highest similarity are boxed (grey). Notably, residues that share sequence similarity with Mus musculus Synaptojanin-1 (SYNJ1) 534-584 fall outside of the SYNJ1 5-phosphatase domain and have not been implicated in its catalytic activity. Local homology between SopB and INPP5B or OCRL failed to be identified by NCBI BLAST. (b) Mutation of lysines 525 and 528 blunt PtdIns(3,4)P2 generation by SopB. Wild-type, K525A, K528A, and C460S SopB amino-terminally tagged with EGFP (bottom panels) were transiently expressed with cPHx3 (inverted grey, main panels). The PM was stained with CellMask before imaging live. Representative confocal sections are presented for each. Note that expression of the K528A mutant led to notable perturbation of cellular morphology including rounding, cortical membrane ‘crumpling’, and blebbing. (c) Normalized intensity of PM cPHx3 from (b) was quantified across n=3 independent experiments (WT, 78 cells; K525A, 81 cells; K528A, 86 cells; C460S, 57 cells). Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). ****P < 0.0001. (d) cPHx3 intensities from K528A- and C460S SopB-transfected cells (red box, panel c) plotted on an expanded axis. Subtle PM-enrichment of cPHx3 was evident in K528A-transfected cells relative to the C460S-transfected mutant. Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). **P = 0.0017. (e,f) Lysine 525 and 528 of SopB are structurally predicted to neighbour the C(X)5R motif. The primary sequence of SopB (residues 1-561) was analysed by RoseTTAFold and the resulting highest ranked model is presented in the surface filling view (grey). The location of lysine 525 and 528 (green) and the C(X)5R motif of SopB (blue) are annotated. Source numerical data are available in source data.

Source data

Extended Data Fig. 6 Generation of PtdIns(3,4)P2 by the S. flexneri effector IpgD does not require class I or class II PI3Ks.

(a) Sequence alignment of the phosphate-binding (P-loop) sequence from SopB (Salmonella enterica) and IpgD (Shigella flexneri). Residue 439 of IpgD encodes the cysteine of the C(X)5R motif. (b) IpgD reduces PM PtdIns(4,5)P2 levels in a C(X)5R-dependent manner. Heterologous expression of EGFP-IpgD (WT or C439S) and PH-PLCδ1 (inverted grey, main panels). The PM was stained with CellMask prior to imaging live. (c) Quantification of PH-PLCδ1 intensity in the PM from (b) across n=3 independent experiments quantifying 82 cells (IpgDWT) and 74 cells (IpgDC439S. Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). ****P < 0.0001. (d) Live cell imaging of heterologously-expressed EGFP-IpgD (WT or C439S) and NES-mCherry-cPHx3 following PM staining with CellMask. Cells were treated with the indicated inhibitors (PI-103, 500 nM; wortmannin, 100 nM) for 20 min prior to and throughout imaging. (e) PM cPHx3 intensity was quantified from (d) across 3 independent trials (IpgDWT, 70 cells; IpgDC439S, 62 cells; IpgDWT(PI-103), 68 cells; IpgDWT(Wortmannin), 76 cells). Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). ****P < 0.0001. (f) PM PtdIns(3,4)P2 synthesis in S. cerevisiae. Galactose-inducible empty vector (control), IpgDWT, or IpgDC439S were induced for 2 hours in yeast that expressed cPHx3. Trypan Blue staining demarcates the cell wall and non-viable yeast. (g) Yeast from (f) were scored for plasmalemmal cPHx3 localization across n=4 independent experiments analysing the following number of yeast: control, 881 cells; IpgDWT, 1165 cells; IpgDC439S, 938 cells. Data are mean ± s.e.m. ****P < 0.0001. Source numerical data are available in source data.

Source data

Extended Data Fig. 7 SopB requires an INPP5E-sensitive plasmalemmal inositide for the generation of PtdIns(3,4)P2.

(a) Inhibition of PtdIns(3,4)P2 generated during invasion by pre-recruitment of INPP5E. HeLa cells expressing mRFP-FKBP-INPP5E (WT or D556A) together with PM-targeted Lyn(11)-FRB and the biosensor cPHx1 (grey) were treated with 1 µM rapamycin for 2 min before addition of wild-type Salmonella (BFP, red) for an additional 10 min. The PM was stained (CellMask) and cells fixed before imaging. Maximum intensity projections (main panels) and confocal sections of the boxed region (bottom panels) are presented. Re-localization of mRFP-FKBP-conjugates to the PM is depicted in the bottom panels. (b) Normalized PM intensity of cPHx1 was quantified from part (a) across n=2 independent trials (INPP5EWT, 90 cells; INPP5ED556A, 85 cells). Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). **P = 0.0019. (c) Comparison of PtdIns(4,5)P2 depletion by chemically induced recruitment of PLCβ3 and INPP5E. Normalized PM intensity of PH-PLCδ1 was quantified from n=3 independent experiments (control, 55 cells; PLCβ3, 56 cells; INPP5E, 74 cells). Data from Fig. 6a,b are re-plotted with INPP5E (acquired in parallel) and support that INPP5E results in a modest PtdIns(4,5)P2-depletion relative to PLCβ3. Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). ***P = 0.0002, *P = 0.0477. (d) PtdIns(4,5)P2-depletion by INPP5E recruitment depends on its catalytic activity. Comparison of HeLa cells co-transfected as in (a) but expressing the biosensor PH-PLCδ1-EGFP. Representative confocal sections are presented following 5-min rapamycin treatment (1 µM) and PM staining with CellMask. (e,f) Chemically induced INPP5E recruitment modestly increases PM PtdIns(4)P. HeLa cells expressing mRFP-FKBP-(control), -PLCβ3, or -INPP5E (wild-type) together with PM-targeted Lyn(11)-FRB and EGFP-2xP4M were imaged live 5 min after treatment with 1 µM rapamycin and staining the PM (CellMask). (e) The normalized PM intensity of 2xP4M was quantified from n=3 independent experiments (control, 55 cells; PLCβ3, 41 cells; INPP5E, 53 cells). Data are trial means ± s.e.m. (foreground) overlaid on cell measurements (grey, background). **P = 0.0020. (f) Representative confocal sections of each channel are presented. Source numerical data are available in source data.

Source data

Extended Data Fig. 8 Time-resolved HPLC–MS analysis of SopB phosphotransferase-phosphatase activities.

(a) Inorganic phosphate release assessed following treatment of liposomes with 2.5 µg recombinant SopBWT or SopBC460S. Liposome composition was (mol%) POPS:PtdIns(4,5)P2 (90:10) and 2.5 nmol PtdIns(4,5)P2 was provided per reaction. At the indicated time points, reactions were terminated by addition of 50 mM NEM. Data are mean ± s.e.m. of duplicate wells. Time 0 min corresponds to a no enzyme control. (b) Separation of PtdIns(3,4)P2 and PtdIns(4,5)P2 regio-isomers in a sample treated with SopBWT. An example HPLC–MS trace derived from LUVs treated with 0.5 µg (71.5 nM) SopBWT for 5 min. Note the appearance of PtdIns(3,4)P2 at ≈20.6 min. (c) Model of the PtdIns(3,4,5)P3 biosensor designed with tandem Bruton’s Tyrosine Kinase (BTK) PH domains (bPHx2). Each component was separated by flexible, serine- and glycine-rich linker sequences. NES, nuclear export signal. (d) Heterologous expression of wild-type SopB induces PM-translocation of bPHx2. Representative confocal micrographs of mCherry-tagged bPHx2 (inverted grey) co-transfected with EGFP-SopBC460S or EGFP-SopBWT. (e) Hypothesized PPIns conversions catalysed by SopB and potential modulation by host phosphatases. In the presence of PtdIns(4,5)P2 in vitro, SopB generates the species PtdIns(3,4,5)P3, PtdIns(3,4)P2, and PtdIns(3)P. The latter species are hypothesized to arise, at least in part, by sequential dephosphorylation of PtdIns(3,4,5)P3. In vivo, Salmonella infection favours the accumulation of PtdIns(3,4)P2 likely due to the high basal activity of host 5-phosphatases (INPP5B, SYNJ1/2, SHIP1/2, and others) that rapidly convert PtdIns(3,4,5)P3 to PtdIns(3,4)P2. It remains unclear if the rapid clearance of PtdIns(3,4)P2 following fission of the Salmonella-containing vacuole from the PM is due to the intrinsic activity of SopB or to the activity of host 4-phosphatases (INPP4A/B). Nonetheless, SopB is sufficient to give rise to PtdIns(3)P in vitro from PtdIns(4,5)P2, arguing for a second –likely minor– pathway to generate this inositide in addition to Vps34-mediated synthesis on the bacterial vacuole (Mallo et. al., 2008). Finally, phosphatidylinositol may arise by direct dephosphorylation of the 4- and 5-positions of PtdIns(4,5)P2 by SopB, or indirectly by dephosphorylation of products of the phosphotransferase reaction. Source numerical data are available in source data.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table

Table 1. Plasmids used in this study. Table 2. Reagents used in this study. Table 3. Antibodies used in this study. Table 4. sgRNA oligonucleotides used in this study.

Supplementary Video 1

Live-cell imaging of cPHx3 (grey, left) expressed in HeLa cells during invasion by RFP-expressing Salmonella (red on left). Frames were acquired at intervals of 30 s and are presented as maximum intensity projections of stacks of serial confocal images with a 12 f.p.s. playback. cPHx3 was pseudo-coloured (right) to ease visualization, according to the inset calibration bar. See corresponding insets in Fig. 1a.

Supplementary Video 2

Time-lapse of fluorescence imaging of cPHx3 (grey, left) expressed during invasion by RFP-expressing Salmonella (red, left) delivered at high (>10) multiplicity of infection. Frames were acquired at intervals of 1 min and single confocal sections are presented at 6 f.p.s. playback. cPHx3 was pseudo-coloured (right) to ease visualization according to the inset calibration bar. See additional examples in Extended Data Fig. 1a.

Supplementary Video 3

Time-lapse of fluorescence imaging of aPHx2 (grey) expressed during invasion by wild-type RFP-expressing Salmonella (red on left). Frames were acquired at intervals of 1 min and are presented at a 6 f.p.s. playback. Merged confocal sections are presented on the left and aPHx2 is pseudo-coloured on the right, as indicated by the inset calibration bar. Additional examples are provided in Extended Data Fig. 1d.

Supplementary Video 4

Confocal time-lapse of NES-iRFP-cPHx3 (grey) during optogenetic activation of EGFP-SopBWT-464-TAG. The 405-nm illumination begins at frame 15 (30 s) and is maintained for the duration of the video (5:29). Captures were made at intervals of 2.3 s and are present at a 60 f.p.s. playback. See the corresponding panels and quantification in Fig. 2.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blot.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blot.

Source Data Extended Data Fig. 3

Unprocessed western blot.

Source Data Extended Data Fig. 4

Unprocessed western blot.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Walpole, G.F.W., Pacheco, J., Chauhan, N. et al. Kinase-independent synthesis of 3-phosphorylated phosphoinositides by a phosphotransferase. Nat Cell Biol 24, 708–722 (2022). https://doi.org/10.1038/s41556-022-00895-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00895-y

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