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.

  • Letter
  • Published:

Anthrax lethal factor cleaves regulatory subunits of phosphoinositide-3 kinase to contribute to toxin lethality

Abstract

Anthrax lethal toxin (LT), produced by Bacillus anthracis, comprises a receptor-binding moiety, protective antigen and the lethal factor (LF) protease1,2. Although LF is known to cleave mitogen-activated protein kinase kinases (MEKs/MKKs) and some variants of the NLRP1 inflammasome sensor, targeting of these pathways does not explain the lethality of anthrax toxin1,2. Here we report that the regulatory subunits of phosphoinositide-3 kinase (PI3K)—p85α (PIK3R1) and p85β (PIK3R2)3,4—are substrates of LF. Cleavage of these proteins in a proline-rich region between their N-terminal Src homology and Bcr homology domains disrupts homodimer formation and impacts PI3K signalling. Mice carrying a mutated p85α that cannot be cleaved by LF show a greater resistance to anthrax toxin challenge. The LF(W271A) mutant cleaves p85α with lower efficiency and is non-toxic to mice but can regain lethality when combined with PI3K pathway inhibitors. We provide evidence that LF targets two signalling pathways that are essential for growth and metabolism and that the disabling of both pathways is likely necessary for lethal anthrax infection.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Alignments and constructs.
Fig. 2: p85 proteins are cleaved by LF.
Fig. 3: LT affects PI3K signalling.
Fig. 4: Mutant p85 mice expressing an LT-resistant variant are more resistant to toxin challenge, and LT-induced lethality in mice requires targeting of both MEK and PI3K pathways.

Similar content being viewed by others

Data availability

Full-length blot scans, data from animal studies and associated statistical analyses can be found within the source data files. Any other data generated during and/or analysed for the current study are available from the corresponding author on reasonable request. Reported structural analogues used by iTASSER 5.1 in creation of the model in Fig. 1 are available in the Protein Data Bank under accession codes 1XA6, 3CXI, 3BYI, 5C5S, 2QV2, 3QIS, 3FK2, 2OSA, 1OW3, 2EE4 and 3CXL. Source data are provided with this paper.

References

  1. Liu, S., Moayeri, M. & Leppla, S. H. Anthrax lethal and edema toxins in anthrax pathogenesis. Trends Microbiol. 22, 317–325 (2014).

    PubMed  PubMed Central  Google Scholar 

  2. Moayeri, M., Leppla, S. H., Vrentas, C., Pomerantsev, A. P. & Liu, S. Anthrax pathogenesis. Annu. Rev. Microbiol. 69, 185–208 (2015).

    CAS  PubMed  Google Scholar 

  3. Mellor, P., Furber, L. A., Nyarko, J. N. & Anderson, D. H. Multiple roles for the p85α isoform in the regulation and function of PI3K signalling and receptor trafficking. Biochem J. 441, 23–37 (2012).

    CAS  PubMed  Google Scholar 

  4. Rathinaswamy, M. K. & Burke, J. E. Class I phosphoinositide 3-kinase (PI3K) regulatory subunits and their roles in signaling and disease. Adv. Biol. Regul. 75, 100657 (2020).

    CAS  PubMed  Google Scholar 

  5. Turk, B. E. Exceptionally selective substrate targeting by the metalloprotease anthrax lethal factor. Adv. Exp. Med. Biol. 1111, 189–203 (2019).

    PubMed  Google Scholar 

  6. Levinsohn, J. L. et al. Anthrax lethal factor cleavage of Nlrp1 is required for activation of the inflammasome. PLoS Pathog. 8, e1002638 (2012).

    PubMed  PubMed Central  Google Scholar 

  7. Vitale, G., Bernardi, L., Napolitani, G., Mock, M. & Montecucco, C. Susceptibility of mitogen-activated protein kinase kinase family members to proteolysis by anthrax lethal factor. Biochem. J. 352(Pt 3), 739–745 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Chopra, A. P., Boone, S. A., Liang, X. & Duesbery, N. S. Anthrax lethal factor proteolysis and inactivation of MAPK kinase. J. Biol. Chem. 278, 9402–9406 (2003).

    CAS  PubMed  Google Scholar 

  9. Turk, B. E. et al. The structural basis for substrate and inhibitor selectivity of the anthrax lethal factor. Nat. Struct. Mol. Biol. 11, 60–66 (2004).

    CAS  PubMed  Google Scholar 

  10. Yang, J. & Zhang, Y. Protein structure and function prediction using I-TASSER. Curr. Protoc. Bioinformatics 52, 5.8.1–5.8.15 (2015).

    Google Scholar 

  11. Roy, A., Kucukural, A. & Zhang, Y. I-TASSER: a unified platform for automated protein structure and function prediction. Nat. Protoc. 5, 725–738 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, Y. I-TASSER server for protein 3D structure prediction. BMC Bioinformatics 9, 40 (2008).

    PubMed  PubMed Central  Google Scholar 

  13. Harpur, A. G. et al. Intermolecular interactions of the p85α regulatory subunit of phosphatidylinositol 3-kinase. J. Biol. Chem. 274, 12323–12332 (1999).

    CAS  PubMed  Google Scholar 

  14. Cheung, L. W. et al. Regulation of the PI3K pathway through a p85α monomer–homodimer equilibrium.eLife 4, e06866 (2015).

    PubMed  PubMed Central  Google Scholar 

  15. Goldberg, A. B., Cho, E., Miller, C. J., Lou, H. J. & Turk, B. E. Identification of a substrate-selective exosite within the metalloproteinase anthrax lethal factor. J. Biol. Chem. 292, 814–825 (2017).

    CAS  PubMed  Google Scholar 

  16. Comer, J. E., Chopra, A. K., Peterson, J. W. & Konig, R. Direct inhibition of T-lymphocyte activation by anthrax toxins in vivo. Infect. Immun. 73, 8275–8281 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Ouyang, W., Torigoe, C., Fang, H., Xie, T. & Frucht, D. M. Anthrax lethal toxin inhibits translation of hypoxia-inducible factor 1α and causes decreased tolerance to hypoxic stress. J. Biol. Chem. 289, 4180–4190 (2014).

    CAS  PubMed  Google Scholar 

  18. Ouyang, W., Guo, P., Fang, H. & Frucht, D. M. Anthrax lethal toxin rapidly reduces c-Jun levels by inhibiting c-Jun gene transcription and promoting c-Jun protein degradation. J. Biol. Chem. 292, 17919–17927 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Haeusgen, W., Herdegen, T. & Waetzig, V. The bottleneck of JNK signaling: molecular and functional characteristics of MKK4 and MKK7. Eur. J. Cell Biol. 90, 536–544 (2011).

    CAS  PubMed  Google Scholar 

  20. Gratton, J. P. et al. Akt down-regulation of p38 signaling provides a novel mechanism of vascular endothelial growth factor-mediated cytoprotection in endothelial cells. J. Biol. Chem. 276, 30359–30365 (2001).

    CAS  PubMed  Google Scholar 

  21. Gutierrez-Uzquiza, A., Arechederra, M., Bragado, P., Aguirre-Ghiso, J. A. & Porras, A. p38α mediates cell survival in response to oxidative stress via induction of antioxidant genes: effect on the p70S6K pathway. J. Biol. Chem. 287, 2632–2642 (2012).

    CAS  PubMed  Google Scholar 

  22. Luyendyk, J. P. et al. Genetic analysis of the role of the PI3K–Akt pathway in lipopolysaccharide-induced cytokine and tissue factor gene expression in monocytes/macrophages. J. Immunol. 180, 4218–4226 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Manning, B. D. & Toker, A. AKT/PKB Signaling: navigating the network. Cell 169, 381–405 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Turke, A. B. et al. MEK inhibition leads to PI3K/AKT activation by relieving a negative feedback on ERBB receptors. Cancer Res. 72, 3228–3237 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang, L. et al. A ERK/RSK-mediated negative feedback loop regulates M-CSF-evoked PI3K/AKT activation in macrophages. FASEB J. 32, 875–887 (2018).

    CAS  PubMed  Google Scholar 

  26. Yu, C. F., Liu, Z. X. & Cantley, L. G. ERK negatively regulates the epidermal growth factor-mediated interaction of Gab1 and the phosphatidylinositol 3-kinase. J. Biol. Chem. 277, 19382–19388 (2002).

    CAS  PubMed  Google Scholar 

  27. Lu, Y. et al. Kinome siRNA-phosphoproteomic screen identifies networks regulating AKT signaling. Oncogene 30, 4567–4577 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Shaw, M., Cohen, P. & Alessi, D. R. The activation of protein kinase B by H2O2 or heat shock is mediated by phosphoinositide 3-kinase and not by mitogen-activated protein kinase-activated protein kinase-2. Biochem. J. 336(Pt 1), 241–246 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Qin, S. & Chock, P. B. Implication of phosphatidylinositol 3-kinase membrane recruitment in hydrogen peroxide-induced activation of PI3K and Akt. Biochemistry 42, 2995–3003 (2003).

    CAS  PubMed  Google Scholar 

  30. Ito, Y., Vogt, P. K. & Hart, J. R. Domain analysis reveals striking functional differences between the regulatory subunits of phosphatidylinositol 3-kinase (PI3K), p85α and p85β. Oncotarget 8, 55863–55876 (2017).

    PubMed  PubMed Central  Google Scholar 

  31. Taniguchi, C. M. et al. Phosphoinositide 3-kinase regulatory subunit p85α suppresses insulin action via positive regulation of PTEN. Proc. Natl Acad. Sci. USA 103, 12093–12097 (2006).

    CAS  PubMed  Google Scholar 

  32. Chagpar, R. B. et al. Direct positive regulation of PTEN by the p85 subunit of phosphatidylinositol 3-kinase. Proc. Natl Acad. Sci. USA 107, 5471–5476 (2010).

    CAS  PubMed  Google Scholar 

  33. Barber, D. F., Alvarado-Kristensson, M., Gonzalez-Garcia, A., Pulido, R. & Carrera, A. C. PTEN regulation, a novel function for the p85 subunit of phosphoinositide 3-kinase. Sci. STKE 2006, pe49 (2006).

    PubMed  Google Scholar 

  34. Fruman, D. A. et al. Hypoglycaemia, liver necrosis and perinatal death in mice lacking all isoforms of phosphoinositide 3-kinase p85α. Nat. Genet. 26, 379–382 (2000).

    CAS  PubMed  Google Scholar 

  35. Fruman, D. A. et al. Impaired B cell development and proliferation in absence of phosphoinositide 3-kinase p85α. Science 283, 393–397 (1999).

    CAS  PubMed  Google Scholar 

  36. Mauvais-Jarvis, F. et al. Reduced expression of the murine p85α subunit of phosphoinositide 3-kinase improves insulin signaling and ameliorates diabetes. J. Clin. Invest. 109, 141–149 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ueki, K. et al. Increased insulin sensitivity in mice lacking p85β subunit of phosphoinositide 3-kinase. Proc. Natl Acad. Sci. USA 99, 419–424 (2002).

    CAS  PubMed  Google Scholar 

  38. Vitale, G. et al. Anthrax lethal factor cleaves the N-terminus of MAPKKs and induces tyrosine/threonine phosphorylation of MAPKs in cultured macrophages. Biochem. Biophys. Res. Commun. 248, 706–711 (1998).

    CAS  PubMed  Google Scholar 

  39. Duesbery, N. S. et al. Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal factor. Science 280, 734–737 (1998).

    CAS  PubMed  Google Scholar 

  40. Hellmich, K. A. et al. Anthrax lethal factor cleaves mouse Nlrp1b in both toxin-sensitive and toxin-resistant macrophages. PLoS ONE 7, e49741 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Bachran, C. & Leppla, S. H. Tumor targeting and drug delivery by anthrax toxin. Toxins 8, 197 (2016).

    PubMed Central  Google Scholar 

  42. Liu, S. et al. Solid tumor therapy by selectively targeting stromal endothelial cells. Proc. Natl Acad. Sci. USA 113, E4079–E4087 (2016).

    CAS  PubMed  Google Scholar 

  43. Cheung, L. W. & Mills, G. B. Targeting therapeutic liabilities engendered by PIK3R1 mutations for cancer treatment. Pharmacogenomics 17, 297–307 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Jaiswal, B. S. et al. Somatic mutations in p85α promote tumorigenesis through class IA PI3K activation. Cancer Cell 16, 463–474 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Wu, H. et al. Regulation of class IA PI 3-kinases: C2 domain–iSH2 domain contacts inhibit p85/p110α and are disrupted in oncogenic p85 mutants. Proc. Natl Acad. Sci. USA 106, 20258–20263 (2009).

    CAS  PubMed  Google Scholar 

  46. Sun, M., Hillmann, P., Hofmann, B. T., Hart, J. R. & Vogt, P. K. Cancer-derived mutations in the regulatory subunit p85α of phosphoinositide 3-kinase function through the catalytic subunit p110α. Proc. Natl Acad. Sci. USA 107, 15547–15552 (2010).

    CAS  PubMed  Google Scholar 

  47. Turturro, S. B. et al. Somatic loss of PIK3R1 may sensitize breast cancer to inhibitors of the MAPK pathway. Breast Cancer Res. Treat. 177, 325–333 (2019).

    CAS  PubMed  Google Scholar 

  48. Park, S. & Leppla, S. H. Optimized production and purification of Bacillus anthracis lethal factor. Protein Expr. Purif. 18, 293–302 (2000).

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Mouse Genetics and Gene Modification Section at the National Institute of Allergy and Infectious Diseases and animal care staff and veterinarians in the Comparative Medicine Branch in National Institutes of Health Building 33. This research was supported by the intramural research programs of the National Institute of Allergy and Infectious Diseases and the National Institute of Dental and Craniofacial Research, National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

M.M. conceived the project. M.M. and M.A.M. designed and performed experiments and analysed the data. M.K.P. and D.O. performed experiments. S.L. designed experiments, performed preliminary mutant LF studies and analysed the data. S.H.L. designed constructs, contributed reagents and analysed the data. R.F. purified proteins and performed mass spectrometry analyses. R.S., T.H.B. and J.S.K. designed and created the knockin mouse model. M.M., M.A.M. and S.H.L. wrote and edited the manuscript with input from all authors.

Corresponding author

Correspondence to Mahtab Moayeri.

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 Mass spectrometry on LF-generated cleavage products.

SH3-BH protein was incubated with LF (molar ratio of 100:1, SH3-BH: LF) overnight at 37 °C. Mass spectrometry on cleavage products shown as F1 and F2 on gel yielded molecular masses labeled above each peak. Top graph shows molecular mass of uncleaved SH3-BH, middle graph shows molecular mass of F1 cleavage product, and bottom graph shows molecular mass of F2 cleavage product.

Source data

Extended Data Fig. 2 LT effects on MKK4 and MKK7.

a, C57BL/6J were injected IP with protective antigen (PA) (35 µg) or LT (35 µg) and spleens harvested at 24 h, lysed and analyzed for MEK1, MKK4 and MKK7 cleavage (sequentially, in order shown, re-probing the same blot, using antibodies reactive to N-termini of the proteins). The MEK1 and MKK7 antibodies were detected with an anti-rabbit secondary antibody tagged with same wavelength IR800 label, while MKK4 was detected with an anti-rabbit antibody tagged with an IR680 dye. In the lower panel both secondary antibodies react with all three primary antibodies, resulting in yellow color where both MKK7 and MEK1 are present (lanes 1,2,5), orange where MKK7 alone is present (lanes 3,4, 6) and a lower migrating (red) MKK4 band present equally in all samples (lanes 1-6). b, BMDMs from C57BL/6J mice were treated with LT (1 µg/mL, 1h and 18 h) and analyzed for MKK4 and MKK3 cleavage. The MKK3 band is not seen in spleen lysates, processed as in (a). In panel a MEK1 and MKK7 bands overlap with each other as well as with MKK4, thus densitometry quantification of MKK4 bands was performed only for panel b (see Supplementary Table 1).

Source data

Extended Data Fig. 3 Signaling and survival consequences of LF cleavage.

a, C57BL/6J bone marrow-derived macrophages (BMDMs) were treated with LT or LTW271A (1 µg/ml) for indicated times, followed by LPS (1 µg/ml, 90 min). p38 inhibitor PH-797804 treatment was at 50 nM for 16 h, then at 100 nM for 1 h prior to LPS stimulation. Lysates were analyzed using antibodies to the N-terminus of MEK1 and re-probed with anti-pAkt (Thr308). b, c, Human foreskin fibroblasts (b) and HT1080 cells (c) were treated with LT (1 µg/ml, 12 h) prior to addition of insulin (200 nM, 45 min) and lysate analyses as in (a). d, Pik3r1GVAA/GVAA (KI) or wild-type (WT) mice were injected with LT (35 µg) and organs harvested at 24 h were analyzed for p85α and MEK1 cleavage. In the color panel, KI or WT BMDMs were treated with LT (1 µg/mL, 12 h). e, BMDMs were pre-treated with LT (1 µg/ml, 2 or 18 h), followed by stimulation with H2O2 (1 mM, 30 min) or insulin (200 nM, 45 min). Lysates were analyzed for pAkt, MEK1 cleavage, and actin. Top blot is initial probe with p-Akt (Thr308) antibody, middle gel is probe of same gel with MEK1 N-terminal antibody and lower gel is a third re-probe with anti β-actin, where β-actin runs with MEK1. f, Organs of mice challenged with mutant toxin LTW271A or LT (35 µg, IP, 18 h) were analyzed for p85α and MEK1 cleavage (g) KI mice and WT littermates were challenged with high dose LT (75 µg, IV) and monitored for survival. P-value comparing curves is 0.8114 by Log-rank test. h, C57BL/6J mice were challenged with LT (100 µg, IP) or LTW271A (100 µg or 250 µg, IP) and monitored for survival. P-value comparing LT (100 µg) to LTW271A (100 µg) is 0.0926, LT (100 µg) to LTW271A (250 µg) is 0.0128, and comparing the two LTW271A curves is 0.7301 (all by Log-rank test). Blue arrows in panels a-e point to cross-reactive bands which serve as internal equal loading controls. For densitometry quantifications see Supplementary Table 1.

Source data

Extended Data Fig. 4 Test of p38 inhibitor function in vivo.

C57BL/6J mice (n=6/group) were treated with either p38 inhibitor Losmapimod (LOS) (25 mg/kg/200 µl by oral gavage) or vehicle at 18 h and 2 h prior to injection of LPS (5 mg/kg/500 µl, IP). TNF-α levels in serum were measured at 2 h post-LPS treatment to assess inhibition of p38 pathway. Serum from two untreated animals was also measured to establish baseline. **** indicates that P-value comparing the vehicle vs. drug treated group is <0.0001 by unpaired t-test (two-tailed).

Source data

Supplementary information

Supplementary Information

Supplementary Table 1 and densitometry.

Reporting Summary

Source data

Source Data Fig. 2

Full-length blot scans.

Source Data Fig. 2

Densitometry readings, enzyme assays and statistical analyses.

Source Data Fig. 3

Full-length blot scans.

Source Data Fig. 4

Full-length blot scans.

Source Data Fig. 4

Animal study data and statistical analyses.

Source Data Extended Data Fig. 1

Full-length blot scans.

Source Data Extended Data Fig. 2

Full-length blot scans.

Source Data Extended Data Fig. 3

Full-length blot scans.

Source Data Extended Data Fig. 3

Animal study data and statistical analyses.

Source Data Extended Data Fig. 4

ELISA data and statistical analyses.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mendenhall, M.A., Liu, S., Portley, M.K. et al. Anthrax lethal factor cleaves regulatory subunits of phosphoinositide-3 kinase to contribute to toxin lethality. Nat Microbiol 5, 1464–1471 (2020). https://doi.org/10.1038/s41564-020-0782-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-0782-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing