Chlamydia causes fragmentation of the Golgi compartment to ensure reproduction (original) (raw)

Nature volume 457, pages 731–735 (2009)Cite this article

Abstract

The obligate intracellular bacterium Chlamydia trachomatis survives and replicates within a membrane-bound vacuole, termed the inclusion, which intercepts host exocytic pathways to obtain nutrients1,2,3. Like many other intracellular pathogens, C. trachomatis has a marked requirement for host cell lipids, such as sphingolipids and cholesterol, produced in the endoplasmic reticulum and the Golgi apparatus4,5,6. However, the mechanisms by which intracellular pathogens acquire host cell lipids are not well understood1,2,3. In particular, no host cell protein responsible for transporting Golgi-derived lipids to the chlamydial inclusions has yet been identified. Here we show that Chlamydia infection in human epithelial cells induces Golgi fragmentation to generate Golgi ministacks surrounding the bacterial inclusion. Ministack formation is triggered by the proteolytic cleavage of the Golgi matrix protein golgin-84. Inhibition of golgin-84 truncation prevents Golgi fragmentation, causing a block in lipid acquisition and maturation of C. trachomatis. Golgi fragmentation by means of RNA-interference-mediated knockdown of distinct Golgi matrix proteins before infection enhances bacterial maturation. Our data functionally connect bacteria-induced golgin-84 cleavage, Golgi ministack formation, lipid acquisition and intracellular pathogen growth. We show that C. trachomatis subverts the structure and function of an entire host cell organelle for its own advantage.

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

Access options

Subscribe to this journal

Receive 51 print issues and online access

$199.00 per year

only $3.90 per issue

Buy this article

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

Additional access options:

Similar content being viewed by others

References

  1. Carabeo, R. A., Mead, D. J. & Hackstadt, T. Golgi-dependent transport of cholesterol to the Chlamydia trachomatis inclusion. Proc. Natl Acad. Sci. USA 100, 6771–6776 (2003)
    Article ADS CAS PubMed PubMed Central Google Scholar
  2. Hackstadt, T., Rockey, D. D., Heinzen, R. A. & Scidmore, M. A. Chlamydia trachomatis interrupts an exocytic pathway to acquire endogenously synthesized sphingomyelin in transit from the Golgi apparatus to the plasma membrane. EMBO J. 15, 964–977 (1996)
    Article CAS PubMed PubMed Central Google Scholar
  3. Scidmore, M. A., Fischer, E. R. & Hackstadt, T. Sphingolipids and glycoproteins are differentially trafficked to the Chlamydia trachomatis inclusion. J. Cell Biol. 134, 363–374 (1996)
    Article CAS PubMed Google Scholar
  4. Hatch, G. M. & McClarty, G. Phospholipid composition of purified Chlamydia trachomatis mimics that of the eucaryotic host cell. Infect. Immun. 66, 3727–3735 (1998)
    CAS PubMed PubMed Central Google Scholar
  5. van Ooij, C. et al. Host cell-derived sphingolipids are required for the intracellular growth of Chlamydia trachomatis . Cell. Microbiol. 2, 627–637 (2000)
    Article CAS PubMed Google Scholar
  6. Wylie, J. L., Hatch, G. M. & McClarty, G. Host cell phospholipids are trafficked to and then modified by Chlamydia trachomatis . J. Bacteriol. 179, 7233–7242 (1997)
    Article CAS PubMed PubMed Central Google Scholar
  7. De Matteis, M. A. & Luini, A. Exiting the Golgi complex. Nature Rev. Mol. Cell Biol. 9, 273–284 (2008)
    Article CAS Google Scholar
  8. Shorter, J. & Warren, G. Golgi architecture and inheritance. Annu. Rev. Cell Dev. Biol. 18, 379–420 (2002)
    Article CAS PubMed Google Scholar
  9. Short, B., Haas, A. & Barr, F. A. Golgins and GTPases, giving identity and structure to the Golgi apparatus. Biochim. Biophys. Acta 1744, 383–395 (2005)
    Article CAS PubMed Google Scholar
  10. Altan-Bonnet, N. et al. Golgi inheritance in mammalian cells is mediated through endoplasmic reticulum export activities. Mol. Biol. Cell 17, 990–1005 (2006)
    Article CAS PubMed PubMed Central Google Scholar
  11. Diao, A. et al. The coiled-coil membrane protein golgin-84 is a novel rab effector required for Golgi ribbon formation. J. Cell Biol. 160, 201–212 (2003)
    Article CAS PubMed PubMed Central Google Scholar
  12. Lowe, M. et al. Cdc2 kinase directly phosphorylates the cis-Golgi matrix protein GM130 and is required for Golgi fragmentation in mitosis. Cell 94, 783–793 (1998)
    Article CAS PubMed Google Scholar
  13. Wang, Y. et al. A direct role for GRASP65 as a mitotically regulated Golgi stacking factor. EMBO J. 22, 3279–3290 (2003)
    Article CAS PubMed PubMed Central Google Scholar
  14. Chiu, R., Novikov, L., Mukherjee, S. & Shields, D. A caspase cleavage fragment of p115 induces fragmentation of the Golgi apparatus and apoptosis. J. Cell Biol. 159, 637–648 (2002)
    Article CAS PubMed PubMed Central Google Scholar
  15. Lowe, M., Lane, J. D., Woodman, P. G. & Allan, V. J. Caspase-mediated cleavage of syntaxin 5 and giantin accompanies inhibition of secretory traffic during apoptosis. J. Cell Sci. 117, 1139–1150 (2004)
    Article CAS PubMed Google Scholar
  16. Mancini, M. et al. Caspase-2 is localized at the Golgi complex and cleaves golgin-160 during apoptosis. J. Cell Biol. 149, 603–612 (2000)
    Article CAS PubMed PubMed Central Google Scholar
  17. Rajalingam, K. et al. Epithelial cells infected with Chlamydophila pneumoniae (Chlamydia pneumoniae) are resistant to apoptosis. Infect. Immun. 69, 7880–7888 (2001)
    Article CAS PubMed PubMed Central Google Scholar
  18. Fan, T. et al. Inhibition of apoptosis in chlamydia-infected cells: blockade of mitochondrial cytochrome c release and caspase activation. J. Exp. Med. 187, 487–496 (1998)
    Article CAS PubMed PubMed Central Google Scholar
  19. Hood, J. L., Brooks, W. H. & Roszman, T. L. Differential compartmentalization of the calpain/calpastatin network with the endoplasmic reticulum and Golgi apparatus. J. Biol. Chem. 279, 43126–43135 (2004)
    Article CAS PubMed Google Scholar
  20. Goll, D. E. et al. The calpain system. Physiol. Rev. 83, 731–801 (2003)
    Article CAS PubMed Google Scholar
  21. Suzuki, K. & Sorimachi, H. A novel aspect of calpain activation. FEBS Lett. 433, 1–4 (1998)
    Article ADS CAS PubMed Google Scholar
  22. Puthenveedu, M. A. et al. GM130 and GRASP65-dependent lateral cisternal fusion allows uniform Golgi-enzyme distribution. Nature Cell Biol. 8, 238–248 (2006)
    Article CAS PubMed Google Scholar
  23. Holthuis, J. C. & Levine, T. P. Lipid traffic: floppy drives and a superhighway. Nature Rev. Mol. Cell Biol. 6, 209–220 (2005)
    Article CAS Google Scholar
  24. Marie, M., Sannerud, R., Avsnes Dale, H. & Saraste, J. Membrane traffic in the secretory pathway: Take the 'A' train: on fast tracks to the cell surface. Cell. Mol. Life Sci. 65, 2859–2874 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  25. Machuy, N. et al. A global approach combining proteome analysis and phenotypic screening with RNA interference yields novel apoptosis regulators. Mol. Cell. Proteomics 4, 44–55 (2005)
    Article CAS PubMed Google Scholar
  26. Heuer, D., Brinkmann, V., Meyer, T. F. & Szczepek, A. J. Expression and translocation of chlamydial protease during acute and persistent infection of the epithelial HEp-2 cells with Chlamydophila (Chlamydia) pneumoniae . Cell. Microbiol. 5, 315–322 (2003)
    Article CAS PubMed Google Scholar

Download references

Acknowledgements

The authors would like to thank A. Greiser, C. Goosmann, B. Laube, M. Wicht and E. Ziska for technical support; M. A. De Matteis for the gift of the GFP–GM130 fusion plasmid; H. P. Hauri for the provision of the anti-GPP130 antibody and helpful discussions; and K. Astrahantseff, S. and J. Heuer, T. Wolff and L. Ogilvie for critically reading the manuscript and helpful suggestions. This work was financially supported by the Senate of Berlin and the BMBF through the RiNA Network.

Author information

Authors and Affiliations

  1. Department of Molecular Biology,,
    Dagmar Heuer, Anette Rejman Lipinski, Nikolaus Machuy, Alexander Karlas, Andrea Wehrens & Thomas F. Meyer
  2. Microscopy Core Facility, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany ,
    Volker Brinkmann
  3. Department of Membrane Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany,
    Frank Siedler

Authors

  1. Dagmar Heuer
    You can also search for this author inPubMed Google Scholar
  2. Anette Rejman Lipinski
    You can also search for this author inPubMed Google Scholar
  3. Nikolaus Machuy
    You can also search for this author inPubMed Google Scholar
  4. Alexander Karlas
    You can also search for this author inPubMed Google Scholar
  5. Andrea Wehrens
    You can also search for this author inPubMed Google Scholar
  6. Frank Siedler
    You can also search for this author inPubMed Google Scholar
  7. Volker Brinkmann
    You can also search for this author inPubMed Google Scholar
  8. Thomas F. Meyer
    You can also search for this author inPubMed Google Scholar

Corresponding author

Correspondence toThomas F. Meyer.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-17 with Legends and Supplementary Table 1 which summarizes truncated golgin-84 expressing cell line, plasmids and primers. (PDF 2543 kb)

Supplementary Movie 1

Supplementary Movie 1 shows GM130-GFP expressing cells infected with C.trachomatis to reveal GA disruption during the infection period. The time lapse covers about 12 h. By following cells during the course of the movie, it is evident that the GA is dispersed during infection. (MOV 3373 kb)

Supplementary Movie 2

Supplementary Movie 2 shows ceramide trafficking in infected cells. BODIPY FL C5-ceramide transport into the inclusion. Time-lapse images of HeLa cells infected with C. trachomatis (MOI = 2) for 26 h. Labelled ceramide was added 1 to 2 minutes after starting image acquisition. The assigned numbers represent the time elapsed. (MOV 2259 kb)

Supplementary Movie 3

Supplementary Movie 3 shows ceramide trafficking in infected cells treated with Z-WEHD-FMK. BODIPY FL C5-ceramide transport into the inclusion is inhibited by treatment with Z-WEHD-FMK. Time-lapse images of HeLa cells infected with C. trachomatis (MOI = 2) treated with inhibitor 9 h p.i. Image acquisition was started at 27 h p.i. and labelled ceramide was added 1 to 2 minutes after starting the acquisition. The assigned numbers represent the time elapsed. (MOV 2566 kb)

PowerPoint slides

Rights and permissions

About this article

Cite this article

Heuer, D., Lipinski, A., Machuy, N. et al. Chlamydia causes fragmentation of the Golgi compartment to ensure reproduction.Nature 457, 731–735 (2009). https://doi.org/10.1038/nature07578

Download citation

This article is cited by

Editorial Summary

Golgi bodies hijacked

Like many other intracellular pathogens Chlamydia trachomatis depends on host lipids for growth. A mechanism by which Chlamydia can acquire its lipids has now been demonstrated in infected HeLa epithelial cells. In the normal cell the Golgi apparatus acts to modify newly synthesized proteins and lipids for distribution within or outside the cell. The intracellular replication of C. trachomatis triggers the breakdown of the Golgi apparatus to generate functionally intact Golgi ministacks via cleavage of the matrix protein golgin-84. The Golgi ministacks, which align around the bacterial inclusion, appear to provide a conduit through which the pathogen can secure a supply of lipids. This work points to mechanistic involvement of inflammatory caspases and calpains, which may be used as novel molecular targets for antichlamydial agents.