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:

VacA generates a protective intracellular reservoir for Helicobacter pylori that is eliminated by activation of the lysosomal calcium channel TRPML1

Abstract

Helicobacter pylori infection is a proven carcinogen for gastric cancer. Its virulence factor vacuolating cytotoxin A (VacA) promotes more severe disease and gastric colonization. VacA, by an unknown mechanism, usurps lysosomal and autophagy pathways to generate a protected reservoir for H. pylori that confers bacterial survival in vitro. Here, we show the existence of a VacA-generated intracellular niche in vivo that protects the bacteria from antibiotic treatment and leads to infection recrudescence after therapy. Furthermore, we report that VacA targets the lysosomal calcium channel TRPML1 to disrupt endolysosomal trafficking and mediate these effects. Remarkably, H. pylori that lack toxigenic VacA colonize enlarged dysfunctional lysosomes in the gastric epithelium of trpml1-null mice, where they are protected from eradication therapy. Furthermore, a small molecule agonist directed against TRPML1 reversed the toxic effects of VacA on endolysosomal trafficking, culminating in the clearance of intracellular bacteria. These results suggest that TRPML1 may represent a therapeutic target for chronic H. pylori infection.

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: VacA generates an intracellular reservoir in vivo leading to bacterial persistence after eradication therapy.
Fig. 2: VacA impairs TRPML1 activity.
Fig. 3: TRPML1 deficiency generates a VacA-like intracellular niche for H. pylori in vivo.
Fig. 4: TRPML1 activation restores VacA-disrupted endolysosomal and autophagy pathways.
Fig. 5: Validation of the VacA–TRPML1 axis in human gastric organoids.
Fig. 6: TRPML1 activation eliminates both the intracellular niche and survival advantage of VacA+ H. pylori.

Similar content being viewed by others

Data availability

Material used in this study is readily available from the authors or from the commercial source. AGS cells and H. pylori 60190 strain are available from the corresponding author.

References

  1. Mégraud, F., Bessède, E. & Varon, C. Helicobacter pylori infection and gastric carcinoma. Clin. Microbiol. Infect. 21, 984–990 (2015).

    Article  Google Scholar 

  2. Hooi, J. K. Y. et al. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology 153, 420–429 (2017).

    Article  Google Scholar 

  3. IARC Helicobacter pylori Working Group. Helicobacter pylori eradication as a strategy for preventing gastric cancer. IARC Working Group Reports, No. 8 (WHO Press, 2014).

  4. Tacconelli, E. et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 18, 318–327 (2018).

    Article  Google Scholar 

  5. Foegeding, N. J., Caston, R. R., McClain, M. S., Ohi, M. D. & Cover, T. L. An overview of Helicobacter pylori VacA toxin biology. Toxins 8, 173 (2016).

    Article  Google Scholar 

  6. Terebiznik, M. R. et al. Helicobacter pylori VacA toxin promotes bacterial intracellular survival in gastric epithelial cells. Infect. Immun. 74, 6599–6614 (2006).

    Article  CAS  Google Scholar 

  7. Terebiznik, M. R. et al. Effect of Helicobacter pylori’s vacuolating cytotoxin on the autophagy pathway in gastric epithelial cells. Autophagy 5, 370–379 (2009).

    Article  CAS  Google Scholar 

  8. Raju, D. et al. Vacuolating cytotoxin and variants in Atg16L1 that disrupt autophagy promote Helicobacter pylori infection in humans. Gastroenterology 142, 1160–1171 (2012).

    Article  CAS  Google Scholar 

  9. Galluzzi, L. et al. Autophagy in malignant transformation and cancer progression. EMBO J. 34, 856–880 (2015).

    Article  CAS  Google Scholar 

  10. Waller-Evans, H. & Lloyd-Evans, E. Regulation of TRPML1 function. Biochem. Soc. Trans. 43, 442–446 (2015).

    Article  CAS  Google Scholar 

  11. Venkatachalam, K., Wong, C. O. & Zhu, M. X. The role of TRPMLs in endolysosomal trafficking and function. Cell Calcium 58, 48–56 (2015).

    Article  CAS  Google Scholar 

  12. Cheng, X., Shen, D., Samie, M. & Xu, H. Mucolipins: intracellular TRPML1-3 channels. FEBS Lett. 584, 2013–2021 (2010).

    Article  CAS  Google Scholar 

  13. Dong, X. et al. PI(3,5)P2 controls membrane trafficking by direct activation of mucolipin Ca2+ release channels in the endolysosome. Nat. Commun. 1, 38 (2010).

    Article  Google Scholar 

  14. Shen, D. et al. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat. Commun. 3, 731 (2012).

    Article  Google Scholar 

  15. Schiffmann, R. et al. Constitutive achlorhydria in mucolipidosis type IV. Proc. Natl Acad. Sci. USA 95, 1207–1212 (1998).

    Article  CAS  Google Scholar 

  16. Venugopal, B. et al. Neurologic, gastric, and opthalmologic pathologies in a murine model of mucolipidosis type IV. Am. J. Hum. Genet. 81, 1070–1083 (2007).

    Article  CAS  Google Scholar 

  17. Vergarajauregui, S., Connelly, P. S., Daniels, M. P. & Puertollano, R. Autophagic dysfunction in mucolipidosis type IV patients. Hum. Mol. Genet. 17, 2723–2737 (2008).

    Article  CAS  Google Scholar 

  18. Chandra, M. et al. A role for the Ca2+ channel TRPML1 in gastric acid secretion, based on analysis of knockout mice. Gastroenterology 140, 857–867 (2011).

    Article  CAS  Google Scholar 

  19. Miller, A. et al. Mucolipidosis type IV protein TRPML1-dependent lysosome formation. Traffic 16, 284–297 (2015).

    Article  CAS  Google Scholar 

  20. Lee, A. et al. A standardized mouse model of Helicobacter pylori infection: introducing the Syndey strain. Gastroenterology 122, 1386–1397 (1997).

    Article  Google Scholar 

  21. Winter, J. A. et al. A role for the vacuolating cytotoxin, VacA, in colonization and Helicobacter pylori-induced metaplasia in the stomach. J. Infect. Dis. 210, 954–963 (2014).

    Article  CAS  Google Scholar 

  22. Salama, N. R., Otto, G., Tompkins, L. & Falkow, S. Vacuolating cytotoxin of Helicobacter pylori plays a role during colonization in a mouse model of infection. Infect. Immun. 69, 730–736 (2001).

    Article  CAS  Google Scholar 

  23. Oertli, M. et al. Helicobacter pylori γ-glutamyl transpeptidase and vacuolating cytotoxin promote gastric persistence and immune tolerance. Proc. Natl Acad. Sci. USA 110, 3047–3052 (2013).

    Article  CAS  Google Scholar 

  24. Veldhuyzen van Zanten, S. J. O., Kolesnikow, T., Leung, V., O’Rourke, J. L. & Lee, A. Gastric transitional zones, areas where Helicobacter treatment fails: results of a treatment trial using the Sydney strain mouse model. Antimicrob. Agents Chemother. 47, 2249–2255 (2003).

    Article  Google Scholar 

  25. Dubois, A. & Borén, T. Helicobacter pylori is invasive and it may be a facultative intracellular organism. Cell. Microbiol. 9, 1108–1116 (2007).

    Article  CAS  Google Scholar 

  26. Necchi, V. et al. Natural history of Helicobacter pylori VacA toxin in human gastric epithelium in vivo: vacuoles and beyond. Sci. Rep. 7, 14526 (2017).

    Article  Google Scholar 

  27. Cao, Q. et al. BK channels alleviate lysosomal storage diseases by providing positive feedback regulation of lysosomal Ca2+ release. Dev. Cell 33, 427–441 (2015).

    Article  CAS  Google Scholar 

  28. Zhong, X. Z., Yang, Y., Sun, X. & Dong, X.-P. Methods for monitoring Ca2+ and ion channels in the lysosome. Cell Calcium 64, 20–28 (2017).

    Article  CAS  Google Scholar 

  29. Cover, T. L., Tummuru, M. K. R., Cao, P., Thompson, S. A. & Blaser, M. J. Divergence of genetic sequences for the vacuolating cytotoxin among Helicobacter pylori strains. J. Biol. Chem. 269, 10566–10573 (1994).

    CAS  PubMed  Google Scholar 

  30. Patel, H. K. et al. Plasma membrane cholesterol modulates cellular vacuolation induced by the Helicobacter pylori vacuolating cytotoxin. Infect. Immun. 70, 4112–4123 (2002).

    Article  CAS  Google Scholar 

  31. McCartney, A. J., Zhang, Y. & Weisman, L. S. Phosphatidylinositol 3,5-bisphosphate: low abundance, high significance. BioEssays 36, 52–64 (2014).

    Article  CAS  Google Scholar 

  32. Chen, C.-C. et al. A small molecule restores function to TRPML1 mutant isoforms responsible for mucolipidosis type IV. Nat. Commun. 5, 1–10 (2014).

    Google Scholar 

  33. Javier Pérez-Victoria, F., Mardones, G. A. & Bonifacino, J. S. Requirement of the human GARP complex for mannose 6-phosphate-receptor-dependent sorting of cathepsin D to lysosomes. Mol. Biol. Cell 19, 2350–2362 (2008).

    Article  Google Scholar 

  34. Coutinho, M. F., Prata, M. J. & Alves, S. Mannose-6-phosphate pathway: a review on its role in lysosomal function and dysfunction. Mol. Genet. Metab. 105, 542–550 (2012).

    Article  CAS  Google Scholar 

  35. Canuel, M., Korkidakis, A., Konnyu, K. & Morales, C. R. Sortilin mediates the lysosomal targeting of cathepsins D and H. Biochem. Biophys. Res. Commun. 373, 292–297 (2008).

    Article  CAS  Google Scholar 

  36. Coutinho, M. F., Prata, M. J. & Alves, S. A shortcut to the lysosome: the mannose-6-phosphate-independent pathway. Mol. Genet. Metab. 107, 257–266 (2012).

    Article  CAS  Google Scholar 

  37. Necchi, V., Sommi, P., Ricci, V. & Solcia, E. In vivo accumulation of Helicobacter pylori products, NOD1, ubiquitinated proteins and proteasome in a novel cytoplasmic structure. PLoS ONE 5, e9716 (2010).

    Article  Google Scholar 

  38. Abe, K. & Puertollano, R. Role of TRP channels in the regulation of the endosomal pathway. Physiology 26, 14–22 (2011).

    Article  CAS  Google Scholar 

  39. Sahoo, N. et al. Gastric acid secretion from parietal cells is mediated by a Ca2+ efflux channel in the tubulovesicle. Dev. Cell 41, 262–273 (2017).

    Article  CAS  Google Scholar 

  40. Remis, N. N. et al. Mucolipin co-deficiency causes accelerated endolysosomal vacuolation of enterocytes and failure-to-thrive from birth to weaning. PLoS Genet. 10, e1004833 (2014).

    Article  Google Scholar 

  41. Xu, H. & Ren, D. Lysosomal physiology. Annu. Rev. Physiol. 77, 57–80 (2015).

    Article  CAS  Google Scholar 

  42. Li, Y., Wandinger-Ness, A., Goldenring, J. R. & Cover, T. L. Clustering and redistribution of late endocytic compartments in response to Helicobacter pylori vacuolating toxin. Mol. Biol. Cell 15, 1946–1959 (2004).

    Article  CAS  Google Scholar 

  43. Palframan, S. L., Kwok, T. & Gabriel, K. Vacuolating cytotoxin A (VacA), a key toxin for Helicobacter pylori pathogenesis. Front. Cell. Infect. Microbiol. 2, 1–9 (2012).

    Article  Google Scholar 

  44. Stauber, T. & Jentsch, T. J. Chloride in vesicular trafficking and function. Annu. Rev. Physiol. 75, 453–477 (2013).

    Article  CAS  Google Scholar 

  45. Kasper, D. et al. Loss of the chloride channel ClC-7 leads to lysosomal storage disease and neurodegeneration. EMBO J. 24, 1079–1091 (2005).

    Article  CAS  Google Scholar 

  46. Bertaux-Skeirik, N. et al. CD44 plays a functional role in Helicobacter pylori-induced epithelial cell proliferation. PLoS Pathog. 11, e1004663 (2015).

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Slaugenhaupt for the Trpml1-deficient mice, S. Blanke for the purified VacA toxin and anti-VacA polyclonal antibody and J. Atherton for the H. pylori SS1 strains. This work was supported by the Canadian Institutes of Health Research (CIHR), the Canadian Association of Gastroenterology (CAG) and the North American Society for Paediatric Gastroenterology, Hepatology and Nutrition Foundation. L.K.G. was supported by a CIHR/CAG/Canadian Crohn’s and Colitis and CIHR/CAG/AbbVie Pharmaceuticals Canada Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

N.L.J. conceived and supervised the study, analysed the data and wrote the manuscript. M.I.C., L.K.G. and A.P. designed, carried out and analysed most of the experiments and wrote the manuscript. S.X., M.A., N.B.-S., J.C., Y.Z., C.O’B. and M.H. assisted with the gastric organoid studies and provided helpful comments for the manuscript. X.Z.Z. and X.D. performed the lysosomal calcium release experiments, analysed the data and provided helpful comments for the manuscript. R.P. provided the human biopsies and I.S. analysed the H. pylori staining and both provided helpful comments for the manuscript. H.W., L.R. and D.J.P. contributed to experimental design and provided helpful comments for the manuscript.

Corresponding author

Correspondence to Nicola L. Jones.

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.

Supplementary information

Supplementary Information

Supplementary Table 1, Supplementary Figures 1–19, uncropped blots and Supplementary Video legends.

Reporting Summary

Supplementary Video 1

SS1 H. pylori is restricted to the glandular lumen in wild-type mice. H. pylori staining of gastric tissue obtained from wild-type infected mice. The video was created by a 3D reconstruction of confocal z-sections acquired each 0.20 µm using a 40× water objective and deconvolved utilizing Volocity software. Similar staining was obtained for all of the wild-type mice (n = 12).

Supplementary Video 2

SS1 H. pylori colonize vacuolar compartments of parietal cells in trpml1−/− mice. H. pylori staining of gastric tissue obtained from Trpml1-deficient infected mice. The video was created by a 3D reconstruction of confocal z-sections acquired each 0.20 µm using a 40× water objective and deconvolved utilizing Volocity software. H. pylori within vacuolar compartments in parietal cells were identified in all of the Trpml1-deficient mice analysed (n = 8).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Capurro, M.I., Greenfield, L.K., Prashar, A. et al. VacA generates a protective intracellular reservoir for Helicobacter pylori that is eliminated by activation of the lysosomal calcium channel TRPML1. Nat Microbiol 4, 1411–1423 (2019). https://doi.org/10.1038/s41564-019-0441-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0441-6

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