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:

A CREB3–ARF4 signalling pathway mediates the response to Golgi stress and susceptibility to pathogens

Abstract

Treatment of cells with brefeldin A (BFA) blocks secretory vesicle transport and causes a collapse of the Golgi apparatus. To gain more insight into the cellular mechanisms mediating BFA toxicity, we conducted a genome-wide haploid genetic screen that led to the identification of the small G protein ADP-ribosylation factor 4 (ARF4). ARF4 depletion preserves viability, Golgi integrity and cargo trafficking in the presence of BFA, and these effects depend on the guanine nucleotide exchange factor GBF1 and other ARF isoforms including ARF1 and ARF5. ARF4 knockdown cells show increased resistance to several human pathogens including Chlamydia trachomatis and Shigella flexneri. Furthermore, ARF4 expression is induced when cells are exposed to several Golgi-disturbing agents and requires the CREB3 (also known as Luman or LZIP) transcription factor, whose downregulation mimics ARF4 loss. Thus, we have uncovered a CREB3–ARF4 signalling cascade that may be part of a Golgi stress response set in motion by stimuli compromising Golgi capacity.

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

Figure 1: Loss of ARF4 provides resistance to several Golgi-disrupting agents.
Figure 2: ARF4 depletion preserves Golgi morphology and protein trafficking in the presence of BFA.
Figure 3: Compensatory upregulation of other ARF family members in ARF4 knockdown cells.
Figure 4: BFA resistance of ARF4-depleted cells depends on ARF1 and GBF1.
Figure 5: Golgi stress causes ARF4 upregulation.
Figure 6: CREB3 mediates ARF4 induction in response to Golgi stress.
Figure 7: ARF4 loss protects against several intracellular pathogens.

Similar content being viewed by others

References

  1. Chiu, V. K. et al. Ras signalling on the endoplasmic reticulum and the Golgi. Nat. Cell Biol. 4, 343–350 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Altan-Bonnet, N., Phair, R. D., Polishchuk, R. S., Weigert, R. & Lippincott-Schwartz, J. A role for Arf1 in mitotic Golgi disassembly, chromosome segregation, and cytokinesis. Proc. Natl Acad. Sci. USA 100, 13314–13319 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Sutterlin, C., Hsu, P., Mallabiabarrena, A. & Malhotra, V. Fragmentation and dispersal of the pericentriolar Golgi complex is required for entry into mitosis in mammalian cells. Cell 109, 359–369 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Yadav, S., Puri, S. & Linstedt, A. D. A primary role for Golgi positioning in directed secretion, cell polarity, and wound healing. Mol. Biol. Cell 20, 1728–1736 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kupfer, A., Louvard, D. & Singer, S. J. Polarization of the Golgi apparatus and the microtubule-organizing center in cultured fibroblasts at the edge of an experimental wound. Proc. Natl Acad. Sci. USA 79, 2603–2607 (1982).

    Article  CAS  PubMed  Google Scholar 

  6. Singleton, V. L., Bohonos, N. & Ullstrup, A. J. Decumbin, a new compound from a species of Penicillium. Nature 181, 1072–1073 (1958).

    Article  CAS  PubMed  Google Scholar 

  7. Shao, R. G., Shimizu, T. & Pommier, Y. Brefeldin A is a potent inducer of apoptosis in human cancer cells independently of p53. Exp. Cell Res. 227, 190–196 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Nojiri, H., Manya, H., Isono, H., Yamana, H. & Nojima, S. Induction of terminal differentiation and apoptosis in human colonic carcinoma cells by brefeldin A, a drug affecting ganglioside biosynthesis. FEBS Lett. 453, 140–144 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Sausville, E. A. et al. Antiproliferative effect in vitro and antitumor activity in vivo of brefeldin A. The Cancer J. Sci. Am. 2, 52–58 (1996).

    CAS  PubMed  Google Scholar 

  10. Anadu, N. O., Davisson, V. J. & Cushman, M. Synthesis and anticancer activity of brefeldin A ester derivatives. J. Med. Chem. 49, 3897–3905 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Donaldson, J. G. & Jackson, C. L. ARF family G proteins and their regulators: roles in membrane transport, development and disease. Nat. Rev. Mol. Cell Biol. 12, 362–375 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. D’Souza-Schorey, C. & Chavrier, P. ARF proteins: roles in membrane traffic and beyond. Nat. Rev. Mol. Cell Biol. 7, 347–358 (2006).

    Article  PubMed  Google Scholar 

  13. Peyroche, A. et al. Brefeldin A acts to stabilize an abortive ARF-GDP-Sec7 domain protein complex: involvement of specific residues of the Sec7 domain. Mol. Cell 3, 275–285 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. D’Souza-Schorey, C., Li, G., Colombo, M. I. & Stahl, P. D. A regulatory role for ARF6 in receptor-mediated endocytosis. Science 267, 1175–1178 (1995).

    Article  PubMed  Google Scholar 

  15. Peters, P. J. et al. Overexpression of wild-type and mutant ARF1 and ARF6: distinct perturbations of nonoverlapping membrane compartments. J. Cell Biol. 128, 1003–1017 (1995).

    Article  CAS  PubMed  Google Scholar 

  16. Cavenagh, M. M. et al. Intracellular distribution of Arf proteins in mammalian cells. Arf6 is uniquely localized to the plasma membrane. J. Biol. Chem. 271, 21767–21774 (1996).

    Article  CAS  PubMed  Google Scholar 

  17. Reiling, J. H. et al. A haploid genetic screen identifies the major facilitator domain containing 2A (MFSD2A) transporter as a key mediator in the response to tunicamycin. Proc. Natl Acad. Sci. USA 108, 11756–11765 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Carette, J. E. et al. Haploid genetic screens in human cells identify host factors used by pathogens. Science 326, 1231–1235 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Carette, J. E. et al. Global gene disruption in human cells to assign genes to phenotypes by deep sequencing. Nature Biotechnol. 29, 542–546 (2011).

    Article  CAS  Google Scholar 

  20. Volpicelli-Daley, L. A., Li, Y., Zhang, C. J. & Kahn, R. A. Isoform-selective effects of the depletion of ADP-ribosylation factors 1-5 on membrane traffic. Mol. Biol. Cell 16, 4495–4508 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nakai, W. et al. ARF1 and ARF4 regulate recycling endosomal morphology and retrograde transport from endosomes to the Golgi apparatus. Mol. Biol. Cell 24, 2570–2581 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kondo, Y. et al. ARF1 and ARF3 are required for the integrity of recycling endosomes and the recycling pathway. Cell Struct. Funct. 37, 141–154 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Claude, A. et al. GBF1: a novel Golgi-associated BFA-resistant guanine nucleotide exchange factor that displays specificity for ADP-ribosylation factor 5. J. Cell Biol. 146, 71–84 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Ooi, C. E., Dell’Angelica, E. C. & Bonifacino, J. S. ADP-Ribosylation factor 1 (ARF1) regulates recruitment of the AP-3 adaptor complex to membranes. J. Cell Biol. 142, 391–402 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shinotsuka, C., Yoshida, Y., Kawamoto, K., Takatsu, H. & Nakayama, K. Overexpression of an ADP-ribosylation factor-guanine nucleotide exchange factor, BIG2, uncouples brefeldin A-induced adaptor protein-1 coat dissociation and membrane tubulation. J. Biol. Chem. 277, 9468–9473 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Alvarez, C., Garcia-Mata, R., Brandon, E. & Sztul, E. COPI recruitment ismodulated by a Rab1b-dependent mechanism. Mol. Biol. Cell 14, 2116–2127 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Teal, S. B., Hsu, V. W., Peters, P. J., Klausner, R. D. & Donaldson, J. G. An activating mutation in ARF1 stabilizes coatomer binding to Golgi membranes. J. Biol. Chem. 269, 3135–3138 (1994).

    CAS  PubMed  Google Scholar 

  28. Santy, L. C. & Casanova, J. E. Activation of ARF6 by ARNO stimulates epithelial cell migration through downstream activation of both Rac1 and phospholipase D. J. Cell Biol. 154, 599–610 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Manolea, F. et al. Arf3 is activated uniquely at the trans-Golgi network by brefeldin A-inhibited guanine nucleotide exchange factors. Mol. Biol. Cell 21, 1836–1849 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bui, Q. T., Golinelli-Cohen, M. P. & Jackson, C. L. Large Arf1 guanine nucleotide exchange factors: evolution, domain structure, and roles in membrane trafficking and human disease. Mol. Genet. Genom. 282, 329–350 (2009).

    Article  CAS  Google Scholar 

  31. Nakagomi, S. et al. A Golgi fragmentation pathway in neurodegeneration. Neurobiol. Dis. 29, 221–231 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Lee, T. H. & Linstedt, A. D. Potential role for protein kinases in regulation of bidirectional endoplasmic reticulum-to-Golgi transport revealed by protein kinase inhibitor H89. Mol. Biol. Cell 11, 2577–2590 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Nickel, W., Helms, J. B., Kneusel, R. E. & Wieland, F. T. Forskolin stimulates detoxification of brefeldin A. J. Biol. Chem. 271, 15870–15873 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Jang, S. Y., Jang, S. W. & Ko, J. Regulation of ADP-ribosylation factor 4 expression by small leucine zipper protein and involvement in breast cancer cell migration. Cancer Lett. 314, 185–197 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Asada, R., Kanemoto, S., Kondo, S., Saito, A. & Imaizumi, K. The signalling from endoplasmic reticulum-resident bZIP transcription factors involved in diverse cellular physiology. J. Biochem. 149, 507–518 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Kondo, S. et al. Activation of OASIS family, ER stress transducers, is dependent on its stabilization. Cell Death Differ. 19, 1939–1949 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Denboer, L. M. et al. JAB1/CSN5 inhibits the activity of Luman/CREB3 by promoting its degradation. Biochim. Biophys. Acta 1829, 921–929 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Heuer, D. et al. Chlamydia causes fragmentation of the Golgi compartment to ensure reproduction. Nature 457, 731–735 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Burnaevskiy, N. et al. Proteolytic elimination of N-myristoyl modifications by the Shigella virulence factor IpaJ. Nature 496, 106–109 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hackstadt, T., Scidmore, M. A. & Rockey, D. D. Lipid metabolism in Chlamydia trachomatis-infected cells: directed trafficking of Golgi-derived sphingolipids to the chlamydial inclusion. Proc. Natl Acad. Sci. USA 92, 4877–4881 (1995).

    Article  CAS  PubMed  Google Scholar 

  41. Elwell, C. A. et al. Chlamydia trachomatis co-opts GBF1 and CERT to acquire host sphingomyelin for distinct roles during intracellular development. PLoS Pathog. 7, e1002198 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gurumurthy, R. K. et al. A loss-of-function screen reveals Ras- and Raf-independent MEK-ERK signaling during Chlamydia trachomatis infection. Sci. Signal. 3, ra21 (2010).

    Article  PubMed  Google Scholar 

  43. Dong, N. et al. Structurally distinct bacterial TBC-like GAPs link Arf GTPase to Rab1 inactivation to counteract host defenses. Cell 150, 1029–1041 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Brunham, R. C. & Rey-Ladino, J. Immunology of Chlamydia infection:implications for a Chlamydia trachomatis vaccine. Nat. Rev. Immunol. 5, 149–161 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Kotloff, K. L. et al. Global burden of Shigella infections: implications for vaccine development and implementation of control strategies. Bull. World Health Organ. 77, 651–666 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Raggo, C. et al. Luman, the cellular counterpart of herpes simplex virus VP16, is processed by regulated intramembrane proteolysis. Mol. Cell Biol. 22, 5639–5649 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. DenBoer, L. M. et al. Luman is capable of binding and activating transcription from the unfolded protein response element. Biochem. Biophys. Res. Commun. 331, 113–119 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Liang, G. et al. Luman/CREB3 induces transcription of the endoplasmic reticulum (ER) stress response protein Herp through an ER stress response element. Mol. Cell Biol. 26, 7999–8010 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen, X., Shen, J. & Prywes, R. The luminal domain of ATF6 senses endoplasmic reticulum (ER) stress and causes translocation of ATF6 from the ER to the Golgi. J. Biol. Chem. 277, 13045–13052 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Nadanaka, S., Okada, T., Yoshida, H. & Mori, K. Role of disulfide bridges formed in the luminal domain of ATF6 in sensing endoplasmic reticulum stress. Mol. Cell Biol. 27, 1027–1043 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Zhang, K. et al. Endoplasmic reticulum stress activates cleavage of CREBH to induce a systemic inflammatory response. Cell 124, 587–599 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Citterio, C. et al. Unfolded protein response and cell death after depletion of brefeldin A-inhibited guanine nucleotide-exchange protein GBF1. Proc. Natl Acad. Sci. USA 105, 2877–2882 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Saenz, J. B. et al. Golgicide A reveals essential roles for GBF1 in Golgi assembly and function. Nat. Chem. Biol. 5, 157–165 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chun, J., Shapovalova, Z., Dejgaard, S. Y., Presley, J. F. & Melancon, P. Characterization of class I and II ADP-ribosylation factors (Arfs) in live cells: GDP-bound class II Arfs associate with the ER–Golgi intermediate compartment independently of GBF1. Mol. Biol. Cell 19, 3488–3500 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Feng, Y. et al. Exo1: a new chemical inhibitor of the exocytic pathway. Proc. Natl Acad. Sci. USA 100, 6469–6474 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Barzilay, E., Ben-Califa, N., Hirschberg, K. & Neumann, D. Uncoupling of brefeldin a-mediated coatomer protein complex-I dissociation from Golgi redistribution. Traffic 6, 794–802 (2005).

    Article  CAS  PubMed  Google Scholar 

  57. Dinter, A. & Berger, E. G. Golgi-disturbing agents. Histochem. Cell Biol. 109, 571–590 (1998).

    Article  CAS  PubMed  Google Scholar 

  58. Zhang, G. F., Driouich, A. & Staehelin, L. A. Effect of monensin on plant Golgi: re-examination of the monensin-induced changes in cisternal architecture and functional activities of the Golgi apparatus of sycamore suspension-cultured cells. J. Cell Sci. 104, 819–831 (1993).

    CAS  PubMed  Google Scholar 

  59. Hicks, S. W. & Machamer, C. E. Golgi structure in stress sensing and apoptosis. Biochim. Biophys. Acta 1744, 406–414 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Oku, M. et al. Novel cis-acting element GASE regulates transcriptional induction by the Golgi stress response. Cell Struct. Funct. 36, 1–12 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Huynh, D. P., Yang, H.T., Vakharia, H., Nguyen, D. & Pulst, S.M. Expansion of the polyQ repeat in ataxin-2 alters its Golgi localization, disrupts the Golgi complex and causes cell death. Human Mol. Genet. 12, 1485–1496 (2003).

    Article  CAS  Google Scholar 

  62. Winslow, A.R. et al. α-Synuclein impairs macroautophagy: implications for Parkinson’s disease. J. Cell Biol. 190, 1023–1037 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Cooper, A. A. et al. α-synuclein blocks ER–Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science 313, 324–328 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Walkley, S.U. & Suzuki, K. Consequences of NPC1 and NPC2 loss of function in mammalian neurons. Biochim. Biophys. Acta 1685, 48–62 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. Gonatas, N. K., Stieber, A. & Gonatas, J. O. Fragmentation of the Golgiapparatus in neurodegenerative diseases and cell death. J. Neurol. Sci. 246, 21–30 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Jehl, S. P., Nogueira, C. V., Zhang, X. & Starnbach, M. N. IFNgamma inhibits the cytosolic replication of Shigella flexneri via the cytoplasmic RNA sensor RIG-I. PLoS Pathog. 8, e1002809 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Gondek, D. C., Olive, A. J., Stary, G. & Starnbach, M. N. CD4+ T cells are necessary and sufficient to confer protection against Chlamydia trachomatis infection in the murine upper genital tract. J. Immunol. 189, 2441–2449 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. van der Velden, A. W., Dougherty, J. T. & Starnbach, M. N. Down-modulation of TCR expression by Salmonella enterica serovar Typhimurium. J. Immunol. 180, 5569–5574 (2008).

    Article  CAS  PubMed  Google Scholar 

  69. Coers, J. et al. Compensatory T cell responses in IRG-deficient mice prevent sustained Chlamydia trachomatis infections. PLoS Pathog. 7, e1001346 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Cohen, L. A. & Donaldson, J. G. Curr. Protoc. Cell Biol. 48, 11–17 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

We thank J. G. Donaldson for providing the pGEX–VHS–GAT construct, R. A. Weinberg for the Calu-1 cell line and D. Kim for help with confocal microscopy. This work was supported by grants from the US National Institutes of Health (NIH; CA103866) and the D. H. Koch Institute for Integrative Cancer Research to D.M.S. D.M.S. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

J.H.R. designed and carried out most of the experiments, analysed data, and wrote the manuscript with input and contributions from all other co-authors. D.M.S. supervised the project, analysed data and edited the manuscript. A.J.O. performed all Chlamydia and Shigella infection assays, edited the manuscript and was supervised by M.N.S. S.S. carried out the pulse-chase labelling and influenza A virus experiments and was supervised by H.L.P. J.E.C. and T.R.B. assisted with haploid genetic screening.

Corresponding authors

Correspondence to Jan H. Reiling or David M. Sabatini.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Effect of BFA and other compounds on ARF4 and ARF5 knockdown cells.

(a) ARF4 knockdown validation of PANC1 and A549 cells infected with ARF4- or control-hairpins. Blots are representative of two or more independent experiments. (b) U251 cells lentivirally-transduced with control or ARF4 shRNAs were seeded in 96-well plates and treated for 3 days with or without indicated drugs before assessing cell viability using the CTG assay. ARF4 knockdown cells are significantly protected against BFA and GCA but not against TM, TG, or A23187 compared with control cells. 12 wells were measured of each genotype, p<10−13 for both shARF4 hairpins compared to control hairpins (student’s t-test). Drug concentrations used and abbreviations are: 30 ng/mL Brefeldin A (BFA), 3 μM Golgicide A (GCA), 500 ng/mL Tunicamycin (TM), 5 nM Thapsigargin (TG) and 1 μg/mL A23187. Two independent experiments were performed. (c) ARF4 or ARF5 depletion in HeLa cells causes resistance and hypersensitivity to BFA, respectively. Cells were left untreated or treated for 3 days with 7.5 ng/mL BFA, and the survival ratio was calculated by counting the number of surviving cells in the presence and absence of BFA. It is likely that the ARF4 antibody used for Western blotting also weakly recognizes ARF5, which is 90% identical to ARF4 (full length ARF4 was used as immunogen for antibody production) as there seems to be a slight ARF4 signal reduction upon ARF5 depletion in this particular cell line. Four wells for the control and shARF4 hairpins were measured and two wells for the individual shARF5 hairpins, p<0.0001 for both ARF4 shRNAs (student’s 2-tailed t-test). (d) knockdown of ARF5 in PANC1 cells makes cells more BFA-sensitive similar as in (c). Treatment duration was 4 days and 30 ng/mL BFA was used. Viability score was calculated as in (c), n = 4 wells for controls and n = 2 wells for each shARF5 hairpin.

Supplementary Figure 2 Protection against BFA treatment upon loss of ARF4.

(a, b) Golgi morphology is preserved in ARF4-depleted HeLa cells but not control cells upon treatment with low BFA concentrations. Shown are representative (two independent experiments) IF images of cells stained for the cis-Golgi marker GM130 and DNA (Hoechst). BFA treatment disrupts the Golgi complex in control but not in ARF4 knockdown cells. (a) Cells left untreated, and (b) cells treated for one hour with 40 ng/mL BFA. (c) Pulse/chase labeling experiment following MHC class I receptor trafficking (revealed by immunoprecipitation with the W6/32 antibody) with or without BFA treatment demonstrates that a functional secretory pathway persists in ARF4 knockdown HeLa cells despite the presence of 50 ng/mL BFA. (d) Short term treatment of A549 cells transduced with indicated hairpins with a high (1 μg/mL) BFA dose disperses the Golgi both in control and ARF4 knockdown cells. Experiment was done twice.

Supplementary Figure 3 Co-knockdowns of ARF4 and ARF5 or ARF3 and their effects on survival following BFA treatment.

(a) ARF4 ARF5 co-depletion makes A549 cells less resistant to BFA in comparison to double-knockdown controls. Cells were treated for 3 days with 20 ng/mL BFA or left untreated, and viable cells were counted in both conditions to calculate the survival ratios. Two wells for each double-knockdown combination and condition were tested. (b) A549 cells simultaneously depleted for ARF3 and ARF4 display the ARF4 single knockdown phenotype indicating a negligible role of ARF3 for BFA resistance upon loss of ARF4. Two wells of each double-knockdown combination and condition were measured. Viability was calculated as in (a). (a, b) Western blotting confirms knockdown of the respective ARFs in the double-knockdown cells.

Supplementary Figure 4 Effect of Golgi stress treatments and CREB3 expression on ARFs and ARF-GEFs.

(a) Shown are quantitative real-time PCR results of BFA treated and vehicle treated cells (treatment duration was 29 hours, and cells were either vehicle-treated or with 20 ng/mL BFA). P-values (student’s 2-tailed t-test) are as follows: for GBF1, p<0.01 and p<0.004 (A549 and HeLa, respectively); BIG1, p<0.05 and p<0.009 (A549 and HeLa, respectively); BIG2, p<0.001 and p<0.004 (A549 and HeLa, respectively). RNA of three wells per genotype and condition was extracted, and two Q real-time PCR reactions were run simultaneously (technical replicates) to derive the average value for the individual genes. (b) HeLa and A549 cells were co-treated with BFA and two different doses of the protein synthesis inhibitor Cycloheximide (CHX) for 29 hours. The experiment was performed twice. (c) Stable Flag-CREB3 and control (Flag- γTubulin) protein-expressing HeLa cells were left untreated or treated for 24 hours with 10 ng/mL BFA before cell lysis, and the lysates tested for ARF isoforms and ER stress marker expression by immunoblotting. Blots are representative of two independent experiments. (d) A549 cells infected with control or CREB3 hairpins were treated for 24 hours with several Golgi stressors and the lysates probed with the indicated antibodies after SDS-PAGE. Experiment was done twice. Note the induction of ARF1 following CREB3 downregulation.

Supplementary Figure 5 Involvement and regulation of CREB3 and S1P in Golgi stress-induced ARF4 induction.

(a) Lentivirally-transduced HeLa cells stably expressing Flag-CREB3 were left untreated or treated for 23 hours with 20 ng/mL BFA, 5 μM GCA, 75 μM Exo1 or 10 μM Monensin before fixation and immunofluorescent staining to assess CREB3 localization and Golgi morphology. Shown are images of two representative experiments. (b) Simultaneous treatment of A549 cells with 10 ng/mL BFA for 28 hours and either of two different serine protease inhibitors (AEBSF or PF-429242) mitigates ARF4 upregulation in a dose-dependent manner. Western blots with independent samples were repeated twice. (c) Pharmacological inhibition of S1P in the presence of several Golgi stress agents decreases ARF4 expression compared with BFA only-treated HeLa cells. Treatment duration was 29 hours and PF-429242 was used at 10 μM. The experiment was done twice.

Supplementary Figure 6 Resistance of ARF4 knockdown cells to C. trachomatis and S. flexneri is mimicked by CREB3 loss of function and depends on ARF5 and GBF1.

(a) HeLa cells transduced with lentiviral control or ARF4 hairpins were infected with C. trachomatis, and cells were lysed 30 or 36 hours after infection. Blots (representative of two independent experiments) were probed with indicated antibodies. Ctr. HSP60 refers to C. trachomatis HSP60. (b) HeLa ARF4 knockdown and control cells were infected with Chlamydia, and 30 hours post-infection cells were fixed and stained for Chlamydia HSP60 (red), GM130 (green) and DNA (Hoechst; blue). Arrows point to Chlamydia-infected cells and the effects on the Golgi apparatus. Representative images of two independent experiments are presented. (c) HeLa double-knockdown cells were infected with C. trachomatis and bacterial progeny tested for their ability to form inclusions in wild type HeLa cells (48 hours post-infection). Statistically significant differences (p<0.05) using one-way ANOVA were observed between ARF4 double-knockdown control cells and ARF4 GBF1 double-knockdown cells; four wells were measured per genotype. The graphs show the mean IFUs/well ± S.D. (d) Immunoblots of cell lysates from HeLa double-knockdown cells used in (c). Two independent experiments were performed yielding similar results. (e) Loss of CREB3 impairs S. flexneri growth (gentamicin protection assay was done 20 hours post-infection) and inhibits C. trachomatis reproduction (analysis done 40 hours post-infection). Both experiments have been repeated twice with 3-4 plates of each genotype, p 0.05 using Student’s t-test or one-way ANOVA test. Displayed are the mean values ± S.D. (f) Western blot analysis of HeLa CREB3 knockdown cells used for S. flexneri and C. trachomatis infection as shown in (e). Cell lysates were derived from both untreated cells and cells treated with 10 ng/mL BFA for 27 hours.

Supplementary Figure 7 Knockdown of ARF1 or GBF1 causes CREB3 translocation into the nucleus.

(a) A549 cells infected with shLUC, shARF1 or shGBF1 lentiviral hairpins were transfected with Flag-CREB3 cDNA to assess CREB3 subcellular localization. BFA treatment of control cells overexpressing CREB3 led to nuclear enrichment of CREB3 thereby validating the transient transfection assay. ShLUC control cells were left untreated or treated with 20 ng/mL BFA for 24 hours, and the remaining images reflect untreated conditions. The Golgi apparatus was visualized with GM130 and nuclei were stained with Hoechst. (b) Assessment of Golgi morphology and knockdown validation of GBF1 knockdown cells using anti-GBF1 antibody. (a, b) IF images are representative of two independent experiments.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1713 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Reiling, J., Olive, A., Sanyal, S. et al. A CREB3–ARF4 signalling pathway mediates the response to Golgi stress and susceptibility to pathogens. Nat Cell Biol 15, 1473–1485 (2013). https://doi.org/10.1038/ncb2865

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2865

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