1887

Abstract

Cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins, produced by numerous Gram-positive pathogens. CDCs depend on host membrane cholesterol for pore formation; some CDCs also require surface-associated human CD59 (hCD59) for binding, conferring specificity for human cells. We purified a recombinant version of a putative CDC encoded in the genome of subsp, tigurilysin (TGY), and used CRISPR/Cas9 to construct hCD59 knockout (KO) HeLa and JEG-3 cell lines. Cell viability assays with TGY on wild-type and hCD59 KO cells showed that TGY is a hCD59-dependent CDC. Two variants of TGY exist among subsp genomes, only one of which is functional. We discovered that a single amino acid change between these two TGY variants determines its activity. Flow cytometry and oligomerization Western blots revealed that the single amino acid difference between the two TGY isoforms disrupts host cell binding and oligomerization. Furthermore, experiments with hCD59 KO cells and cholesterol-depleted cells demonstrated that TGY is fully dependent on both hCD59 and cholesterol for activity, unlike other known hCD59-dependent CDCs. Using full-length CDCs and toxin constructs differing only in the binding domain, we determined that having hCD59 dependence leads to increased lysis efficiency, conferring a potential advantage to organisms producing hCD59-dependent CDCs.

Funding
This study was supported by the:
  • NIH/NIAID (Award AI155476)
    • Principle Award Recipient: AdamJ Ratner
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License.
Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.001393
2023-09-13
2024-04-30
Loading full text...

Full text loading...

/deliver/fulltext/micro/169/9/mic001393.html?itemId=/content/journal/micro/10.1099/mic.0.001393&mimeType=html&fmt=ahah

References

  1. Gonzalez MR, Bischofberger M, Pernot L, van der Goot FG, Frêche B. Bacterial pore-forming toxins: the (w)hole story?. Cell Mol Life Sci 2008; 65:493–507 [View Article] [PubMed]
    [Google Scholar]
  2. Stanley SA, Raghavan S, Hwang WW, Cox JS. Acute infection and macrophage subversion by Mycobacterium tuberculosis require a specialized secretion system. Proc Natl Acad Sci 2003; 100:13001–13006 [View Article] [PubMed]
    [Google Scholar]
  3. Hensler ME, Liu GY, Sobczak S, Benirschke K, Nizet V et al. Virulence role of group B Streptococcus beta-hemolysin/cytolysin in a neonatal rabbit model of early-onset pulmonary infection. J Infect Dis 2005; 191:1287–1291 [View Article] [PubMed]
    [Google Scholar]
  4. Orihuela CJ, Gao G, Francis KP, Yu J, Tuomanen EI. Tissue-specific contributions of pneumococcal virulence factors to pathogenesis. J Infect Dis 2004; 190:1661–1669 [View Article] [PubMed]
    [Google Scholar]
  5. Geny B, Popoff MR. Bacterial protein toxins and lipids: pore formation or toxin entry into cells. Biol Cell 2006; 98:667–678 [View Article] [PubMed]
    [Google Scholar]
  6. Lim JH, Stirling B, Derry J, Koga T, Jono H et al. Tumor suppressor CYLD regulates acute lung injury in lethal Streptococcus pneumoniae infections. Immunity 2007; 27:349–360 [View Article] [PubMed]
    [Google Scholar]
  7. Inoshima I, Inoshima N, Wilke GA, Powers ME, Frank KM et al. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat Med 2011; 17:1310–1314 [View Article] [PubMed]
    [Google Scholar]
  8. Ratner AJ, Aguilar JL, Shchepetov M, Lysenko ES, Weiser JN. Nod1 mediates cytoplasmic sensing of combinations of extracellular bacteria. Cell Microbiol 2007; 9:1343–1351 [View Article] [PubMed]
    [Google Scholar]
  9. Collier RJ. Membrane translocation by anthrax toxin. Mol Aspects Med 2009; 30:413–422 [View Article] [PubMed]
    [Google Scholar]
  10. Los FCO, Randis TM, Aroian RV, Ratner AJ. Role of pore-forming toxins in bacterial infectious diseases. Microbiol Mol Biol Rev 2013; 77:173–207 [View Article] [PubMed]
    [Google Scholar]
  11. Tweten RK. Cholesterol-dependent cytolysins, a family of versatile pore-forming toxins. Infect Immun 2005; 73:6199–6209 [View Article] [PubMed]
    [Google Scholar]
  12. Hotze EM, Le HM, Sieber JR, Bruxvoort C, McInerney MJ et al. Identification and characterization of the first cholesterol-dependent cytolysins from Gram-negative bacteria. Infect Immun 2013; 81:216–225 [View Article]
    [Google Scholar]
  13. Olofsson A, Hebert H, Thelestam M. The projection structure of perfringolysin O (Clostridium perfringens theta-toxin). FEBS Lett 1993; 319:125–127 [View Article] [PubMed]
    [Google Scholar]
  14. Bhakdi S, Tranum-Jensen J, Sziegoleit A. Mechanism of membrane damage by streptolysin-O. Infect Immun 1985; 47:52–60 [View Article] [PubMed]
    [Google Scholar]
  15. Rossjohn J, Feil SC, McKinstry WJ, Tweten RK, Parker MW. Structure of a cholesterol-binding, thiol-activated cytolysin and a model of its membrane form. Cell 1997; 89:685–692 [View Article] [PubMed]
    [Google Scholar]
  16. Polekhina G, Giddings KS, Tweten RK, Parker MW. Insights into the action of the superfamily of cholesterol-dependent cytolysins from studies of intermedilysin. Proc Natl Acad Sci 2005; 102:600–605 [View Article] [PubMed]
    [Google Scholar]
  17. Wickham SE, Hotze EM, Farrand AJ, Polekhina G, Nero TL et al. Mapping the intermedilysin-human CD59 receptor interface reveals a deep correspondence with the binding site on CD59 for complement binding proteins C8alpha and C9. J Biol Chem 2011; 286:20952–20962 [View Article] [PubMed]
    [Google Scholar]
  18. Lawrence SL, Gorman MA, Feil SC, Mulhern TD, Kuiper MJ et al. Structural basis for receptor recognition by the human CD59-responsive cholesterol-dependent cytolysins. Structure 2016; 24:1488–1498 [View Article] [PubMed]
    [Google Scholar]
  19. Shatursky O, Heuck AP, Shepard LA, Rossjohn J, Parker MW et al. The mechanism of membrane insertion for a cholesterol-dependent cytolysin: a novel paradigm for pore-forming toxins. Cell 1999; 99:293–299 [View Article] [PubMed]
    [Google Scholar]
  20. Shepard LA, Shatursky O, Johnson AE, Tweten RK. The mechanism of pore assembly for a cholesterol-dependent cytolysin: formation of a large prepore complex precedes the insertion of the transmembrane beta-hairpins. Biochemistry 2000; 39:10284–10293 [View Article] [PubMed]
    [Google Scholar]
  21. Czajkowsky DM, Hotze EM, Shao Z, Tweten RK. Vertical collapse of a cytolysin prepore moves its transmembrane beta-hairpins to the membrane. EMBO J 2004; 23:3206–3215 [View Article] [PubMed]
    [Google Scholar]
  22. Giddings KS, Zhao J, Sims PJ, Tweten RK. Human CD59 is a receptor for the cholesterol-dependent cytolysin intermedilysin. Nat Struct Mol Biol 2004; 11:1173–1178 [View Article] [PubMed]
    [Google Scholar]
  23. Gelber SE, Aguilar JL, Lewis KLT, Ratner AJ. Functional and phylogenetic characterization of vaginolysin, the human-specific cytolysin from Gardnerella vaginalis. J Bacteriol 2008; 190:3896–3903 [View Article] [PubMed]
    [Google Scholar]
  24. Tabata A, Matsumoto A, Fujimoto A, Ohkura K, Ikeda T et al. Dual functions of discoidinolysin, a cholesterol-dependent cytolysin with N-terminal discoidin domain produced from Streptococcus mitis strain Nm-76. J Oral Microbiol 2022; 14:2105013 [View Article] [PubMed]
    [Google Scholar]
  25. Zbinden A, Mueller NJ, Tarr PE, Eich G, Schulthess B et al. Streptococcus tigurinus, a novel member of the Streptococcus mitis group, causes invasive infections. J Clin Microbiol 2012; 50:2969–2973 [View Article] [PubMed]
    [Google Scholar]
  26. Zbinden A, Mueller NJ, Tarr PE, Spröer C, Keller PM et al. Streptococcus tigurinus sp. nov., isolated from blood of patients with endocarditis, meningitis and spondylodiscitis. Int J Syst Evol Microbiol 2012; 62:2941–2945 [View Article] [PubMed]
    [Google Scholar]
  27. Bourassa L, Clarridge JE. Clinical significance and characterization of Streptococcus tigurinus isolates in an adult population. J Clin Microbiol 2015; 53:3574–3579 [View Article] [PubMed]
    [Google Scholar]
  28. Kanamori H, Kakuta R, Yano H, Suzuki T, Gu Y et al. A case of culture-negative endocarditis due to Streptococcus tigurinus. J Infect Chemother 2015; 21:138–140 [View Article] [PubMed]
    [Google Scholar]
  29. Shahi I, Llaneras CN, Perelman SS, Torres VJ, Ratner AJ. Genome-wide CRISPR-Cas9 screen does not identify host factors modulating Streptococcus agalactiae β-hemolysin/cytolysin-induced cell death. Microbiol Spectr 2022; 10:e0218621 [View Article] [PubMed]
    [Google Scholar]
  30. Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat Methods 2014; 11:783–784 [View Article] [PubMed]
    [Google Scholar]
  31. Rampersaud R, Planet PJ, Randis TM, Kulkarni R, Aguilar JL et al. Inerolysin, a cholesterol-dependent cytolysin produced by Lactobacillus iners. J Bacteriol 2011; 193:1034–1041 [View Article] [PubMed]
    [Google Scholar]
  32. Diene SM, François P, Zbinden A, Entenza JM, Resch G. Comparative genomics analysis of Streptococcus tigurinus strains identifies genetic elements specifically and uniquely present in highly virulent strains. PLoS One 2016; 11:e0160554 [View Article] [PubMed]
    [Google Scholar]
  33. Johnson S, Brooks NJ, Smith RAG, Lea SM, Bubeck D. Structural basis for recognition of the pore-forming toxin intermedilysin by human complement receptor CD59. Cell Rep 2013; 3:1369–1377 [View Article] [PubMed]
    [Google Scholar]
  34. Ramachandran R, Heuck AP, Tweten RK, Johnson AE. Structural insights into the membrane-anchoring mechanism of a cholesterol-dependent cytolysin. Nat Struct Biol 2002; 9:823–827 [View Article] [PubMed]
    [Google Scholar]
  35. Soltani CE, Hotze EM, Johnson AE, Tweten RK. Structural elements of the cholesterol-dependent cytolysins that are responsible for their cholesterol-sensitive membrane interactions. Proc Natl Acad Sci 2007; 104:20226–20231 [View Article] [PubMed]
    [Google Scholar]
  36. Farrand AJ, LaChapelle S, Hotze EM, Johnson AE, Tweten RK. Only two amino acids are essential for cytolytic toxin recognition of cholesterol at the membrane surface. Proc Natl Acad Sci 2010; 107:4341–4346 [View Article] [PubMed]
    [Google Scholar]
  37. Giddings KS, Johnson AE, Tweten RK. Redefining cholesterol’s role in the mechanism of the cholesterol-dependent cytolysins. Proc Natl Acad Sci 2003; 100:11315–11320 [View Article] [PubMed]
    [Google Scholar]
  38. Soltani CE, Hotze EM, Johnson AE, Tweten RK. Specific protein-membrane contacts are required for prepore and pore assembly by a cholesterol-dependent cytolysin. J Biol Chem 2007; 282:15709–15716 [View Article] [PubMed]
    [Google Scholar]
  39. Zilnyte M, Venclovas Č, Zvirbliene A, Pleckaityte M. The cytolytic activity of vaginolysin strictly depends on cholesterol and is potentiated by human CD59. Toxins 2015; 7:110–128 [View Article] [PubMed]
    [Google Scholar]
  40. Tabata A, Ohkura K, Ohkubo Y, Tomoyasu T, Ohkuni H et al. The diversity of receptor recognition in cholesterol-dependent cytolysins. Microbiol Immunol 2014; 58:155–171 [View Article] [PubMed]
    [Google Scholar]
  41. Kadioglu A, Weiser JN, Paton JC, Andrew PW. The role of Streptococcus pneumoniae virulence factors in host respiratory colonization and disease. Nat Rev Microbiol 2008; 6:288–301 [View Article] [PubMed]
    [Google Scholar]
  42. Weiser JN, Ferreira DM, Paton JC. Streptococcus pneumoniae: transmission, colonization and invasion. Nat Rev Microbiol 2018; 16:355–367 [View Article] [PubMed]
    [Google Scholar]
  43. Petrova MI, Reid G, Vaneechoutte M, Lebeer S. Lactobacillus iners: friend or foe?. Trends Microbiol 2017; 25:182–191 [View Article] [PubMed]
    [Google Scholar]
  44. Thapa R, Ray S, Keyel PA. Interaction of macrophages and cholesterol-dependent cytolysins: the impact on immune response and cellular survival. Toxins 2020; 12:531 [View Article] [PubMed]
    [Google Scholar]
  45. Johnstone BA, Joseph R, Christie MP, Morton CJ, McGuiness C et al. Cholesterol-dependent cytolysins: the outstanding questions. IUBMB Life 2022; 74:1169–1179 [View Article] [PubMed]
    [Google Scholar]
  46. Tomoyasu T, Matsumoto A, Takao A, Tabata A, Nagamune H. A simple method to differentiate three classes of cholesterol-dependent cytolysins. J Microbiol Methods 2023; 207:106696 [View Article] [PubMed]
    [Google Scholar]
  47. Shewell LK, Day CJ, Jen FE-C, Haselhorst T, Atack JM et al. All major cholesterol-dependent cytolysins use glycans as cellular receptors. Sci Adv 2020; 6:eaaz4926 [View Article] [PubMed]
    [Google Scholar]
  48. Shewell LK, Harvey RM, Higgins MA, Day CJ, Hartley-Tassell LE et al. The cholesterol-dependent cytolysins pneumolysin and streptolysin O require binding to red blood cell glycans for hemolytic activity. Proc Natl Acad Sci 2014; 111:E5312–20 [View Article] [PubMed]
    [Google Scholar]
  49. Subramanian K, Neill DR, Malak HA, Spelmink L, Khandaker S et al. Pneumolysin binds to the mannose receptor C type 1 (MRC-1) leading to anti-inflammatory responses and enhanced pneumococcal survival. Nat Microbiol 2019; 4:62–70 [View Article] [PubMed]
    [Google Scholar]
  50. Boyd CM, Parsons ES, Smith RAG, Seddon JM, Ces O et al. Disentangling the roles of cholesterol and CD59 in intermedilysin pore formation. Sci Rep 2016; 6:38446 [View Article] [PubMed]
    [Google Scholar]
  51. Jost BH, Lucas EA, Billington SJ, Ratner AJ, McGee DJ. Arcanolysin is a cholesterol-dependent cytolysin of the human pathogen Arcanobacterium haemolyticum. BMC Microbiol 2011; 11:239 [View Article] [PubMed]
    [Google Scholar]
  52. Drabavicius G, Daelemans D. Intermedilysin cytolytic activity depends on heparan sulfates and membrane composition. PLoS Genet 2021; 17:e1009387 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.001393
Loading
/content/journal/micro/10.1099/mic.0.001393
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error