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:

Typhoid toxin causes neuropathology by disrupting the blood–brain barrier

Abstract

Typhoid fever, primarily caused by Salmonella Typhi, can result in severe life-threatening complications such as encephalopathy. Here we elucidate the mechanisms by which typhoid toxin, a unique virulence factor of S. Typhi, mediates the neuropathology associated with typhoid fever. Utilizing mice engineered to have specific tissues protected from toxin action and an in vitro model of the blood–brain barrier (BBB), we demonstrate that, rather than direct action on neuronal or glial cells, typhoid toxin causes neuropathology by disrupting the BBB. Intravenous tracer studies confirmed significant BBB permeability changes following toxin exposure, an effect we found to be mediated by typhoid toxin’s CdtB catalytic subunit. We demonstrate that corticosteroids are effective at mitigating BBB disruption in vivo, supporting their use for managing typhoid fever neurological complications. Our data reveal mechanistic insight into how typhoid toxin causes encephalopathy and suggest targeted therapeutic interventions to alleviate the severe neurological manifestations of typhoid fever.

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: Preventing typhoid toxin neuronal and glial cell targeting does not prevent intoxication.
Fig. 2: Typhoid toxin disrupts the blood–brain barrier.
Fig. 3: Encephalopathy is associated with typhoid toxin disruption of the blood–brain barrier.
Fig. 4: Direct administration of typhoid toxin into the central nervous system causes no pathologies.
Fig. 5: Typhoid toxin disruption of the blood–brain barrier is mediated by its CdtB subunit.
Fig. 6: Typhoid toxin directly targets blood–brain barrier endothelial cells.

Similar content being viewed by others

Data availability

All data are provided within the main paper, Supplementary Information and Source data files.

References

  1. Voetsch, A. et al. FoodNet estimate of the burden of illness caused by nontyphoidal Salmonella infections in the United States. Clin. Infect. Dis. 38, S127–S134 (2004).

    PubMed  Google Scholar 

  2. Parry, C. & Threlfall, E. Antimicrobial resistance in typhoidal and nontyphoidal salmonellae. Curr. Opin. Infect. Dis. 21, 531–538 (2008).

    PubMed  CAS  Google Scholar 

  3. Crump, J. & Mintz, E. Global trends in typhoid and paratyphoid fever. Clin. Infect. Dis. 50, 241–246 (2010).

    PubMed  Google Scholar 

  4. Parry, C., Hien, T. T., Dougan, G., White, N. & Farrar, J. Typhoid fever. N. Engl. J. Med. 347, 1770–1782 (2002).

    PubMed  CAS  Google Scholar 

  5. Raffatellu, M., Wilson, R., Winter, S. & Bäumler, A. Clinical pathogenesis of typhoid fever. J. Infect. Dev. Ctries 2, 260–266 (2008).

    PubMed  Google Scholar 

  6. Huang, D. & DuPont, H. Problem pathogens: extra-intestinal complications of Salmonella enterica serotype Typhi infection. Lancet Infect. Dis. 5, 341–348 (2005).

    PubMed  Google Scholar 

  7. Osuntokun, B., Bademosi, O., Ogunremi, K. & Wright, S. Neuropsychiatric manifestations of typhoid fever in 959 patients. Arch. Neurol. 27, 7–13 (1972).

    PubMed  CAS  Google Scholar 

  8. Stuart, B. & Pullen, R. Typhoid; clinical analysis of 360 cases. Arch. Intern. Med. 78, 629–661 (1946).

    CAS  Google Scholar 

  9. Panwar, A. & Taksande, A. Typhoid encephalopathy in children: review article. MedLife Clin. 2, 75–76 (2020).

    Google Scholar 

  10. Mellon, G. et al. Encephalitis in a traveller with typhoid fever: efficacy of corticosteroids. J. Travel Med. https://doi.org/10.1093/jtm/tax2063 (2017).

  11. Esa, H., Norazlah, B., Hameed, A., Ding, C. & Wahab, A. Typhoid fever presenting as acute psychosis in a young adult: case report and literature review of typhoid psychosis. Trop. Biomed. 38, 192–195 (2021).

    PubMed  CAS  Google Scholar 

  12. Murillo-Cerda, F. & Martínez-Reséndez, M. Typhoid encephalopathy as a neuropsychiatric manifestation of salmonellosis. Rev. Gastroenterol. Mex. 87, 259–260 (2022).

    CAS  Google Scholar 

  13. Galán, J. Typhoid toxin provides a window into typhoid fever and the biology of Salmonella Typhi. Proc. Natl Acad. Sci. USA 113, 6338–6344 (2016).

    PubMed  PubMed Central  Google Scholar 

  14. Fowler, C. et al. Emerging insights into the biology of typhoid toxin. Curr. Opin. Microbiol. 35, 70–77 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  15. Song, J., Gao, X. & Galan, J. E. Structure and function of the Salmonella Typhi chimaeric A(2)B(5) typhoid toxin. Nature 499, 350–354 (2013).

    PubMed  PubMed Central  CAS  Google Scholar 

  16. Spano, S., Ugalde, J. E. & Galan, J. E. Delivery of a Salmonella Typhi exotoxin from a host intracellular compartment. Cell Host Microbe 3, 30–38 (2008).

    PubMed  CAS  Google Scholar 

  17. Fowler, C., Stack, G., Jiao, X., Lara-Tejero, M. & Galán, J. Alternate subunit assembly diversifies the function of a bacterial toxin. Nat. Commun. https://doi.org/10.1038/s41467-41019-11592-41460 (2019).

  18. Yang, Y.-A. et al. In vivo tropism of Salmonella Typhi toxin to cells expressing a multiantennal glycan receptor. Nat. Microbiol. 3, 155–163 (2018).

    PubMed  CAS  Google Scholar 

  19. Deng, L. et al. Host adaptation of a bacterial toxin from the human pathogen Salmonella Typhi. Cell 159, 1290–1299 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  20. Chou, H. et al. Inactivation of CMP-N-acetylneuraminic acid hydroxylase occurred prior to brain expansion during human evolution. Proc. Natl Acad. Sci. USA 99, 11736–11741 (2002).

    PubMed  PubMed Central  CAS  Google Scholar 

  21. Chou, H. H. et al. A mutation in human CMP-sialic acid hydroxylase occurred after the Homo-Pan divergence. Proc. Natl Acad. Sci. USA 95, 11751–11756 (1998).

    PubMed  PubMed Central  CAS  Google Scholar 

  22. Friedrich, G. & Soriano, P. Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice. Genes Dev. 5, 1513–1523 (1991).

    PubMed  CAS  Google Scholar 

  23. Casola, S. Mouse models for miRNA expression: the ROSA26 locus. Methods Mol. Biol. 667, 145–163 (2010).

    PubMed  CAS  Google Scholar 

  24. Lewandoski, M., Meyers, E. & Martin, G. Analysis of Fgf8 gene function in vertebrate development. Cold Spring Harb. Symp. Quant. Biol. 62, 159–168 (1997).

    PubMed  CAS  Google Scholar 

  25. Tronche, F. et al. Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat. Genet. 23, 99–103 (1999).

    PubMed  CAS  Google Scholar 

  26. Lendahl, U., Zimmerman, L. B. & McKay, R. D. CNS stem cells express a new class of intermediate filament protein. Cell 60, 585–595 (1990).

    PubMed  CAS  Google Scholar 

  27. Mignone, J. L., Kukekov, V., Chiang, A. S., Steindler, D. & Enikolopov, G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469, 311–324 (2004).

    PubMed  CAS  Google Scholar 

  28. Zhu, Y. et al. Ablation of NF1 function in neurons induces abnormal development of cerebral cortex and reactive gliosis in the brain. Genes Dev. 15, 859–876 (2001).

    PubMed  PubMed Central  CAS  Google Scholar 

  29. Obermeier, B., Daneman, R. & Ransohoff, R. M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 19, 1584–1596 (2013).

    PubMed  PubMed Central  CAS  Google Scholar 

  30. Sweeney, M. D., Zhao, Z., Montagne, A., Nelson, A. R. & Zlokovic, B. V. Blood-brain barrier: from physiology to disease and back. Physiol. Rev. 99, 21–78 (2019).

    PubMed  CAS  Google Scholar 

  31. Nguyen, T. et al. The role of 9-O-acetylated glycan receptor moieties in the typhoid toxin binding and intoxication. PLoS Pathog. 16, e1008336 (2020).

    PubMed  PubMed Central  CAS  Google Scholar 

  32. Nag, S. Blood-brain barrier permeability using tracers and immunohistochemistry. Methods Mol. Med. 89, 133–144 (2003).

    PubMed  CAS  Google Scholar 

  33. Keep, R. F. et al. Vascular disruption and blood-brain barrier dysfunction in intracerebral hemorrhage. Fluids Barriers CNS 11, 18 (2014).

    PubMed  PubMed Central  Google Scholar 

  34. Atkinson, A. J. Jr. Intracerebroventricular drug administration. Transl. Clin. Pharm. 25, 117–124 (2017).

    Google Scholar 

  35. Huang, X., Hussain, B. & Chang, J. Peripheral inflammation and blood-brain barrier disruption: effects and mechanisms. CNS Neurosci. Ther. 27, 36–47 (2021).

    PubMed  CAS  Google Scholar 

  36. Sun, Y., Koyama, Y. & Shimada, S. Inflammation from peripheral organs to the brain: how does systemic inflammation cause neuroinflammation? Front. Aging Neurosci. 14, 903455 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  37. Proctor, J., Zang, K., Wang, D., Wang, R. & Reichardt, L. Vascular development of the brain requires β8 integrin expression in the neuroepithelium. J. Neurosci. 25, 9940–9948 (2005).

    PubMed  PubMed Central  CAS  Google Scholar 

  38. Kisanuki, Y. Y. et al. Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo. Dev. Biol. 230, 230–242 (2001).

    PubMed  CAS  Google Scholar 

  39. Brown, T. et al. A microfluidic model of human brain (μHuB) for assessment of blood brain barrier. Bioeng. Transl. Med. 4, e10126 (2019).

    PubMed  PubMed Central  Google Scholar 

  40. Lara-Tejero, M. & Galán, J. E. A bacterial toxin that controls cell cycle progression as a deoxyribonuclease I-like protein. Science 290, 354–357 (2000).

    PubMed  CAS  Google Scholar 

  41. Haghjoo, E. & Galan, J. E. Salmonella Typhi encodes a functional cytolethal distending toxin that is delivered into host cells by a bacterial-internalization pathway. Proc. Natl Acad. Sci. USA 101, 4614–4619 (2004).

    PubMed  PubMed Central  CAS  Google Scholar 

  42. Hoffman, S. L. et al. Reduction of mortality in chloramphenicol-treated severe typhoid fever by high-dose dexamethasone. N. Engl. J. Med. 310, 82–88 (1984).

    PubMed  CAS  Google Scholar 

  43. Punjabi, N. H. et al. Treatment of severe typhoid fever in children with high dose dexamethasone. Pediatr. Infect. Dis. J. 7, 598–600 (1988).

    PubMed  CAS  Google Scholar 

  44. Chisti, M. J. et al. High-dose intravenous dexamethasone in the management of diarrheal patients with enteric fever and encephalopathy. Southeast Asian J. Trop. Med. Public Health 40, 1065–1073 (2009).

    PubMed  CAS  Google Scholar 

  45. Nain, Z. High dose dexamethasone in complicated typhoid fever: what is the evidence? Indian J. Pediatr. 90, 1162 (2023).

    PubMed  Google Scholar 

  46. Background document: the diagnosis, treatment and prevention of typhoid fever. Document number WHO/V&B/03.07 (World Health Organization, 2003); https://iris.who.int/handle/10665/370492

  47. Patabendige, A. & Janigro, D. The role of the blood-brain barrier during neurological disease and infection. Biochem. Soc. Trans. 51, 613–626 (2023).

    PubMed  PubMed Central  CAS  Google Scholar 

  48. Yang, R. et al. Blood-brain barrier integrity damage in bacterial meningitis: the underlying link, mechanisms, and therapeutic targets. Int. J. Mol. Sci. https://doi.org/10.3390/ijms24032852 (2023).

  49. Peng, X., Luo, Z., He, S., Zhang, L. & Li, Y. Blood-brain barrier disruption by lipopolysaccharide and sepsis-associated encephalopathy. Front. Cell Infect. Microbiol. 11, 768108 (2021).

    PubMed  PubMed Central  CAS  Google Scholar 

  50. Jiao, X. et al. Generation and characterization of typhoid toxin-neutralizing human monoclonal antibodies. Infect. Immun. https://doi.org/10.1128/iai.00292-20 (2020).

  51. Sasaki, Y. et al. Canonical NF-κB activity, dispensable for B cell development, replaces BAFF-receptor signals and promotes B cell proliferation upon activation. Immunity 24, 729–739 (2006).

    PubMed  CAS  Google Scholar 

  52. Paxinos, G. & Franklin, K. B. J. The Mouse Brain in Stereotaxic Coordinates 2nd edn (Academic Press, 2001).

  53. Taylor, Z. V., Khand, B., Porgador, A., Monsonego, A. & Eremenko, E. An optimized intracerebroventricular injection of CD4+ T cells into mice. STAR Protoc. 2, 100725 (2021).

    PubMed  PubMed Central  CAS  Google Scholar 

  54. Kumar, M. & Nerurkar, V. R. In vitro and in vivo blood-brain barrier models to study West Nile virus pathogenesis. Methods Mol. Biol. 1435, 103–113 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  55. Boyé, K. et al. Endothelial Unc5B controls blood-brain barrier integrity. Nat. Commun. 13, 1169 (2022).

    PubMed  PubMed Central  Google Scholar 

  56. Kozler, P., Marešová, D. & Pokorný, J. Determination of brain water content by dry/wet weight measurement for the detection of experimental brain edema. Physiol. Res. 71, S277–S283 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Hansen, Yale University, for the generous gift of the hCMEC/D3 cell line; M. Shanabrough, Yale University, for help with the electron microscopy experiments; and T. Nottoli at the Yale Genome Editing Center for assistance in the construction of the Cmahfl mouse. This work was supported by NIH Grants R01AI114618 to J.E.G.

Author information

Authors and Affiliations

Authors

Contributions

H.Z. conducted most of the experiments. J.C. assisted with BBB experiments. G.S. constructed the Cmahfl mouse. A.K.A. assisted with the electron microscopy experiments. M.L.-T. provided intellectual input in the project and assisted in the design of the Cmahfl mouse model. T.L.H. provided intellectual input in the project in all aspects related to the BBB and brain pathology. J.E.G. conceived and directed the project. H.Z. and J.E.G wrote the paper with comments from all authors.

Corresponding author

Correspondence to Jorge E. Galán.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Christian Schwerk and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Survival and weight changes in different mouse lines following administration of various doses of typhoid toxin.

Survival (a, c and e) or weight change (b, d and f) of the indicated mice after receiving 0.1, 0.5, or 1 μg of typhoid toxin via the retro-orbital route. n = 4 animals per category. Weight change data are normalized using the initial weight as 100% and represent the mean ± SD for 4 independent measurements.

Source data

Extended Data Fig. 2 Typhoid toxin directly targets blood-brain barrier endothelial cells.

(a) Coronal sections of Cmahfl or Cmahfl/Tek-cre mouse brains (as indicated), 24 and 48 h after receiving 5 μg of Alexa 488-labeled typhoid toxin via the retro-orbital route. (b) Representative fluorescence images of ex vivo toxin binding to brain sections of Cmahfl or Cmahfl/Tek-cre mouse brains (as indicated). Brain sections were fixed and stained with 1 μg Alexa 488-conjugated typhoid toxin (green). Whole brain fluorescence images taken with a 4 x objective were stitched together using the Fusion software package. Scale bar, 1500 μm.

Extended Data Fig. 3 In vivo tropism of typhoid toxin in different mouse lines.

Immunofluorescence images of brain sections of indicated mice 1 h after receiving 5 µg Alexa 488-labeled typhoid toxin (green) via the retro-orbital route. The left panels show whole brain images taken with a 4 x objective and stitched together using the Fusion software package. DAPI (blue) was used to visualized nuclei. The right panels show insets of close-up areas (20 x objective) of the brain sections on the left. To visualize blood vessel endothelial cells, brain sections were stained with a primary rat antibody against mouse CD31 followed by Alexa 568-labeled goat anti-rat antibody (red).

Extended Data Fig. 4 Schematic of the microfluidic device-based 3D BBB in vitro model.

Immortalized human cerebral microvascular endothelial cells hCMEC/D3 are seeded in the apical chamber to form a 3D single-layer vessel, with medium flow to mimic sheer stress from blood flow in vivo to induce expression of tight junction proteins. The slits between the apical chamber and the inner and outer chambers are 3 μm wide and allow measurement of transendothelial electric resistance through the electrode ports (labeled as ‘E’ in the schematic). The device is symmetrical and allows a biological replicate.

Extended Data Fig. 5 Typhoid toxin disrupts the blood-brain barrier tight junctions.

Immunofluorescence images of hCMEC/D3 human brain microvascular endothelial cells cultured in a 3D in vitro system after growing in medium with or without 10 nM wild-type typhoid toxin (a) or its derivatives assembled with its CdtBH160Q (b) or PltAE133A (c) catalytic mutant subunits. Cells were stained with primary mouse antibody against Zonula occludens (ZO-1) followed by Alexa Fluor 488-labeled goat anti-mouse antibody (green) and DAPI (blue) to stain the nuclei. The left panels show a low magnification (4 x objective) image of the entire device and the right panels an inset of a close-up area (20 x objective).

Extended Data Fig. 6 Typhoid toxin induces cell death and activates caspase-3/7 through the Dnase activity of the CdtB subunit.

(a) Cell death and (b) relative caspase-3/7 activity in hCMEC/D3 cells following intoxication with wild-type typhoid toxin or toxoid containing the catalytically inactive CdtBH160Q mutant. Toxin and toxoid concentrations (nM) are indicated. Cell death was quantified by flow cytometry of propidium iodide (PI)-stained cells at the specified time points post-intoxication. Caspase-3/7 activity was measured using the Caspase-Glo® 3/7 luminescence assay, with values normalized to vehicle treated control cells. Data represent the mean ± SD of four biological replicates.

Source data

Extended Data Fig. 7 Effect of dexamethasone on the typhoid toxin-mediated disruption of the blood-brain barrier.

C57BL/6 mice received a sublethal dose of typhoid toxin (500 ng) delivered via the retro-orbital route. Starting 4 days post-intoxication, animals received subcutaneously either dexamethasone (3 μg/g body weight) or eluent (PBS) daily until the end of the experiment. Survival (a), weight change (b), relative Evans blue tracer leakage across the blood-brain barrier (c), or water content of the brain (d) were monitored over time in all the animals. Weight change data were normalized using initial weight as 100% and represent the mean ± SD for measurements in 6 animals (b). For the measurement of Evans blue tracer leakage (c) or the brain water content (d), SD are shown (n = 4 animals per data point before day 4, and n = 3 per category per data point afterwards). Statistically analysis was performed using unpaired two-tailed Student’s t test; *, p < 0.05. The p values were as follows: (c) 0.033453, 0.014107, 0.044075, and 0.026804; (d) 0.027178, and 0.041198. (e) mRNA levels of proinflammatory cytokines in mouse brains 9 days after receiving typhoid toxin (500 ng) through the retro-orbital route. Starting 4 days post-intoxication, animals received subcutaneously either dexamethasone (3 μg/g body weight) or eluent (PBS) daily (n = 5 animals per group). Values were normalized using Gapdh and represent fold induction over mock-treated animal controls. The mean ± SD and p values of the indicated comparison (unpaired two-tailed Student’s t test) are shown.

Source data

Extended Data Fig. 8

Map of plasmid pSB8922 used for the construction of mice with floxed Cmah for tissue specific expression.

Extended Data Table 1 Neurological symptoms in animals following toxin administration

Supplementary information

Supplementary Information

Supplementary Table 1.

Reporting Summary

Supplementary Movie 1

Human brain microvascular hCMEC/D3 cells grown on a microfluidic device to model the blood–brain barrier. Cells were stained with primary mouse antibody against Zonula occludens (ZO-1), followed by Alexa 488-labelled goat anti-mouse antibody (green) and DAPI (blue) to visualize nuclei.

Supplementary Movie 2

Human brain microvascular hCMEC/D3 cells grown on a microfluidic device 24 h after typhoid toxin treatment. Cells were stained with primary mouse antibody against ZO-1, followed by Alexa 488-labelled goat anti-mouse antibody (green) and DAPI (blue) to visualize nuclei.

Supplementary Movie 3

Human brain microvascular hCMEC/D3 cells grown on a microfluidic device 48 h after typhoid toxin treatment. Cells were stained with primary mouse antibody against ZO-1, followed by Alexa 488-labelled goat anti-mouse antibody (green) and DAPI (blue) to visualize nuclei.

Supplementary Movie 4

C57BL/6 mouse exhibiting lethargy 4 days post administration of 5 µg of typhoid toxin via the retro-orbital route.

Supplementary Movie 5

C57BL/6 mouse exhibiting ataxia 4 days post administration of 5 µg of typhoid toxin via the retro-orbital route.

Supplementary Movie 6

C57BL/6 mice exhibiting hyperexcitability 4 days post administration of 5 µg of typhoid toxin via the retro-orbital route.

Supplementary Movie 7

C57BL/6 mouse exhibiting seizure-like behaviour 4 days post administration of 5 µg of typhoid toxin via the retro-orbital route.

Source data

Source Data Figs. 1–6 and Extended Data Figs. 1, 6 and 7.

Raw data and statistical analysis for Figs. 1–6 and Extended Data Figs. 1, 6 and 7.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, H., Catarino, J., Stack, G. et al. Typhoid toxin causes neuropathology by disrupting the blood–brain barrier. Nat Microbiol 10, 1340–1351 (2025). https://doi.org/10.1038/s41564-025-02000-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-025-02000-z

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