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:

ER-shaping atlastin proteins act as central hubs to promote flavivirus replication and virion assembly

Abstract

Flaviviruses, including dengue virus and Zika virus, extensively remodel the cellular endomembrane network to generate replication organelles that promote viral genome replication and virus production. However, it remains unclear how these membranes and associated cellular proteins act during the virus cycle. Here, we show that atlastins (ATLs), a subset of ER resident proteins involved in neurodegenerative diseases, have dichotomous effects on flaviviruses—with ATL2 depletion leading to replication organelle defects, and ATL3 depletion to changes in virus production pathways. We characterized non-conserved functional domains in ATL paralogues and show that the ATL interactome is profoundly reprogrammed following dengue virus infection. Screen analysis confirmed non-redundant ATL functions and identified a specific role for ATL3, and its interactor ARF4, in vesicle trafficking and virion maturation. Our data identify ATLs as central hubs targeted by flaviviruses to establish their replication organelle and to achieve efficient virion maturation and secretion.

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: Impact of atlastin depletion on flavivirus replication.
Fig. 2: Atlastins associate with DV proteins.
Fig. 3: Mapping of functional domains of ATLs involved in the DV replication cycle.
Fig. 4: Comparative ATL interaction networks.
Fig. 5: ATL3 and its interactor ARF4 play important roles in DV maturation.
Fig. 6: ATL3 depletion alters retrograde transport.

Similar content being viewed by others

Data Availability

UniprotKB accession codes of all protein groups and proteins identified by mass spectrometry are provided in each respective Supplementary Table 8 and were extracted from UniprotKB (Human; release 2015_08 including isoforms and unreviewed sequences). Protein sequences of DV-2 16681 strain (P29990) were extracted from UniprotKB. The MS-based proteomics data were deposited at the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the PRIDE partner repository with the data set identifier PXD014639 and PXD014640. For the evolutionary tree analysis, all accession codes can be found in Supplementary Table 7. The remainder of the data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. World Health, O. Dengue vaccine: WHO position paper, July 2016 — recommendations. Vaccine 35, 1200–1201 (2017).

    Article  Google Scholar 

  2. Wikan, N. & Smith, D. R. Zika virus: history of a newly emerging arbovirus. Lancet Infect. Dis. 16, e119–e126 (2016).

    Article  PubMed  Google Scholar 

  3. Neufeldt, C. J., Cortese, M., Acosta, E. G. & Bartenschlager, R. Rewiring cellular networks by members of the Flaviviridae family. Nat. Rev. Microbiol. 16, 125–142 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cortese, M. et al. Ultrastructural characterization of zika virus replication factories. Cell Rep. 18, 2113–2123 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Welsch, S. et al. Composition and three-dimensional architecture of the dengue virus replication and assembly sites. Cell Host Microbe 5, 365–375 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gillespie, L. K., Hoenen, A., Morgan, G. & Mackenzie, J. M. The endoplasmic reticulum provides the membrane platform for biogenesis of the flavivirus replication complex. J. Virol. 84, 10438–10447 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Goyal, U. & Blackstone, C. Untangling the web: mechanisms underlying ER network formation. Biochim. Biophys. Acta 1833, 2492–2498 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Powers, R. E., Wang, S., Liu, T. Y. & Rapoport, T. A. Reconstitution of the tubular endoplasmic reticulum network with purified components. Nature 543, 257–260 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, S., Tukachinsky, H., Romano, F. B. & Rapoport, T. A. Cooperation of the ER-shaping proteins atlastin, lunapark, and reticulons to generate a tubular membrane network. eLife 5, e18605 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Hu, J. & Rapoport, T. A. Fusion of the endoplasmic reticulum by membrane-bound GTPases. Semin. Cell Dev. Biol. 60, 105–111 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Zhang, H. & Hu, J. Shaping the endoplasmic reticulum into a social network. Trends Cell Biol. 26, 934–943 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Hu, J. et al. A class of dynamin-like GTPases involved in the generation of the tubular ER network. Cell 138, 549–561 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Orso, G. et al. Homotypic fusion of ER membranes requires the dynamin-like GTPase atlastin. Nature 460, 978–983 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Rismanchi, N., Soderblom, C., Stadler, J., Zhu, P. P. & Blackstone, C. Atlastin GTPases are required for Golgi apparatus and ER morphogenesis. Hum. Mol. Genet. 17, 1591–1604 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhao, G. et al. Mammalian knock out cells reveal prominent roles for atlastin GTPases in ER network morphology. Exp. Cell Res. 349, 32–44 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Salinas, S., Proukakis, C., Crosby, A. & Warner, T. T. Hereditary spastic paraplegia: clinical features and pathogenetic mechanisms. Lancet Neurol. 7, 1127–1138 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Fischer, D. et al. A novel missense mutation confirms ATL3 as a gene for hereditary sensory neuropathy type 1. Brain 137, e286 (2014).

    Article  PubMed  Google Scholar 

  18. Kornak, U. et al. Sensory neuropathy with bone destruction due to a mutation in the membrane-shaping atlastin GTPase 3. Brain 137, 683–692 (2014).

    Article  PubMed  Google Scholar 

  19. Zhao, X. et al. Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia. Nat. Genet. 29, 326–331 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Behrendt, L., Kurth, I. & Kaether, C. A disease causing ATLASTIN 3 mutation affects multiple endoplasmic reticulum-related pathways. Cellul. Mol. Life Sci. 76, 1433–1445 (2019).

    Article  CAS  Google Scholar 

  21. Hu, X., Wu, F., Sun, S., Yu, W. & Hu, J. Human atlastin GTPases mediate differentiated fusion of endoplasmic reticulum membranes. Protein Cell 6, 307–311 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Klemm, R. W. et al. A conserved role for atlastin GTPases in regulating lipid droplet size. Cell Rep. 3, 1465–1475 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ortiz Sandoval, C. & Simmen, T. Rab proteins of the endoplasmic reticulum: functions and interactors. Biochem. Soc. Trans. 40, 1426–1432 (2012).

    Article  PubMed  CAS  Google Scholar 

  24. .Pawar, S., Ungricht, R., Tiefenboeck, P., Leroux, J. C. & Kutay, U. Efficient protein targeting to the inner nuclear membrane requires Atlastin-dependent maintenance of ER topology. eLife 6, e28202 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Liang, J. R., Lingeman, E., Ahmed, S. & Corn, J. E. Atlastins remodel the endoplasmic reticulum for selective autophagy. J. Cell Bio.l 217, 3354–3367 (2018).

    Article  CAS  Google Scholar 

  26. Krols, M. et al. Sensory-neuropathy-causing mutations in ATL3 cause aberrant ER membrane tethering. Cell Rep. 23, 2026–2038 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Leonetti, M. D., Sekine, S., Kamiyama, D., Weissman, J. S. & Huang, B. A scalable strategy for high-throughput GFP tagging of endogenous human proteins. Proc. Natl Acad. Sci. USA 113, E3501–E3508 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Kudelko, M. et al. Class II ADP-ribosylation factors are required for efficient secretion of dengue viruses. J. Biol. Chem. 287, 767–777 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

  31. Mazelova, J. et al. Ciliary targeting motif VxPx directs assembly of a trafficking module through Arf4. Embo J. 28, 183–192 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. Cortese, M. et al. Reciprocal effects of fibroblast growth factor receptor signaling on dengue virus replication and virion production. Cell Rep. 27, 2579–2592 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Yu, I. M. et al. Association of the pr peptides with dengue virus at acidic pH blocks membrane fusion. J. Virol. 83, 12101–12107 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Moravec, R., Conger, K. K., D’Souza, R., Allison, A. B. & Casanova, J. E. BRAG2/GEP100/IQSec1 interacts with clathrin and regulates alpha5beta1 integrin endocytosis through activation of ADP ribosylation factor 5 (Arf5). J. Biol. Chem. 287, 31138–31147 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chia, P. Z., Gasnereau, I., Lieu, Z. Z. & Gleeson, P. A. Rab9-dependent retrograde transport and endosomal sorting of the endopeptidase furin. J. Cell Sci. 124, 2401–2413 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mallet, W. G. & Maxfield, F. R. Chimeric forms of furin and TGN38 are transported with the plasma membrane in the trans-Golgi network via distinct endosomal pathways. J Cell. Biol. 146, 345–359 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lennemann, N. J. & Coyne, C. B. Dengue and Zika viruses subvert reticulophagy by NS2B3-mediated cleavage of FAM134B. Autophagy 13, 322–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Khaminets, A. et al. Regulation of endoplasmic reticulum turnover by selective autophagy. Nature 522, 354–358 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Friedman, J. R., Dibenedetto, J. R., West, M., Rowland, A. A. & Voeltz, G. K. Endoplasmic reticulum-endosome contact increases as endosomes traffic and mature. Mol. Biol. Cell 24, 1030–1040 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Di Mattia, T., Tomasetto, C. & Alpy, F. Faraway, so close! Functions of endoplasmic reticulum-endosome contacts. Biochim. Biophys. Acta Mol. Cell Biol. Lipids https://doi.org/10.1016/j.bbalip.2019.06.016 (2019).

    Article  CAS  Google Scholar 

  43. Iglesias, N. G. et al. Dengue virus uses a non-canonical function of the host GBF1-Arf-COPI system for capsid protein accumulation on lipid droplets. Traffic 16, 962–977 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Faust, J. E. et al. The Atlastin C-terminal tail is an amphipathic helix that perturbs the bilayer structure during endoplasmic reticulum homotypic fusion. J. Biol. Chem. 290, 4772–4783 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wu, F., Hu, X., Bian, X., Liu, X. & Hu, J. Comparison of human and Drosophila atlastin GTPases. Protein Cell 6, 139–146 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Friebe, P., Boudet, J., Simorre, J. P. & Bartenschlager, R. Kissing-loop interaction in the 3′ end of the hepatitis C virus genome essential for RNA replication. J. Virol. 79, 380–392 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Fischl, W. & Bartenschlager, R. High-throughput screening using dengue virus reporter genomes. Methods Mol. Biol. 1030, 205–219 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Munster, M. et al. A reverse genetics system for zika virus based on a simple molecular cloning strategy. Viruses 10, E368 (2018).

    Article  PubMed  CAS  Google Scholar 

  49. Kuri, T., Habjan, M., Penski, N. & Weber, F. Species-independent bioassay for sensitive quantification of antiviral type I interferons. Virol. J. 7, 50 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Vermeire, J. et al. Quantification of reverse transcriptase activity by real-time PCR as a fast and accurate method for titration of HIV, lenti- and retroviral vectors. PLoS ONE 7, e50859 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Pizzato, M. et al. A one-step SYBR Green I-based product-enhanced reverse transcriptase assay for the quantitation of retroviruses in cell culture supernatants. J. Virol. Methods 156, 1–7 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Edgar, R. C. MUSCLE: a multiple sequence alignment method with reduced time and space complexity. BMC Bioinform. 5, 113 (2004).

    Article  CAS  Google Scholar 

  54. Ronquist, F. & Huelsenbeck, J. P. MrBayes 3: Bayesian phylogenetic inference under mixed models. Bioinformatics 19, 1572–1574 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Stamatakis, A. RAxML-VI-HPC: maximum likelihood-based phylogenetic analyses with thousands of taxa and mixed models. Bioinformatics 22, 2688–2690 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Le, S. Q. & Gascuel, O. An improved general amino acid replacement matrix. Mol. Biol. Evol. 25, 1307–1320 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Doerflinger, S. Y. et al. Membrane alterations induced by nonstructural proteins of human norovirus. PLoS Pathog. 13, e1006705 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Lee, J. Y. et al. Spatiotemporal coupling of the hepatitis C virus replication cycle by creating a lipid droplet-proximal membranous replication compartment. Cell Rep. 27, 3602–3617 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Griffiths, G., Simons, K., Warren, G. & Tokuyasu, K. T. Immunoelectron microscopy using thin, frozen sections: application to studies of the intracellular transport of Semliki Forest virus spike glycoproteins. Methods Enzymol. 96, 466–485 (1983).

    Article  CAS  PubMed  Google Scholar 

  60. Scaturro, P., Cortese, M., Chatel-Chaix, L., Fischl, W. & Bartenschlager, R. Dengue virus non-structural protein 1 modulates infectious particle production via interaction with the structural proteins. PLoS Pathog. 11, e1005277 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Scaturro, P. et al. An orthogonal proteomic survey uncovers novel Zika virus host factors. Nature 561, 253–257 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).

    Article  CAS  PubMed  Google Scholar 

  63. Holze, C. et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat. Immunol. 19, 130–140 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Meth. 13, 731–740 (2016).

    Article  CAS  Google Scholar 

  65. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Ruggieri, L. Chatel-Chaix and M. Joyce for constructive scientific discussions and valuable comments. We also thank U. Haselmann, M. Bartenschlager, I. Paron, A. Piras and S. Kallis for excellent technical support. We acknowledge the Electron Microscopy Core Facility at Heidelberg University and the Infectious Diseases Imaging Platform (IDIP) at the Center for Integrative Infectious Disease Research, Heidelberg, Germany, for expert assistance and access to their equipment as well as the Department of Proteomics and Signal Transduction of the Max-Planck Institute of Biochemistry for continuous and generous support. We thank the European Virus Archive (EVAg, France) for provision of ZV strains, J. Schmidt-Chanasit for providing WNV strains, F. Weber for providing the RVFV isolate, D. Trono for providing the lentiviral transduction system and M. Stanifer for providing the VSVg constructs. This work was supported by grants from the Deutsche Forschungsgemeinschaft (SFB1129, TP11 and BA1505/8-1, both to R.B., and TRR179 and TP11 to A.P.) and the ERC (StG iVIP 331339, CoG ProDAP 817798) to A.P. C.J.N. was supported in part by a European Molecular Biology Organization (EMBO) Long-Term Fellowship (ALTF 466-2016).

Author information

Authors and Affiliations

Authors

Contributions

C.J.N. designed the concept of the project, carried out the majority of the experiments, interpreted the results and wrote the manuscript. M.C. conceived and carried out experiments, and interpreted the results. P.S. performed and analysed mass spectrometry data. J.W. performed and interpreted evolutionary analysis and sequence comparisons. B.C. produced endogenously tagged cell lines. T.M. performed some siRNA experiments. K.T. produced knockout cell lines. O.O. provided reagents and advice for STED microscopy. A.P. contributed to the analysis of mass spectrometry data as well as critical discussions. R.B. contributed to the concept of the study, interpretation of the results and manuscript writing, supervised the project and secured funding.

Corresponding authors

Correspondence to Christopher J. Neufeldt or Ralf Bartenschlager.

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 ER protein depletion in virus infection.

a, Phylogenetic analysis of metazoan ATL proteins collected from a subset of publicly available metazoan predicted proteomes. ATL sequences were aligned with MUSCLE and manually trimmed into a 494-site alignment. Phylogenies were reconstructed, and node support values were calculated using MrBayes for posterior probability and RAxML for maximum likelihood and presented as inset (MrBayes/RAxML). The MrBayes tree topology is shown. Scale bar: number of estimated substitutions per site. For genomes used see Supplementary Table 7 b-h, A549 cells were transduced with constructs encoding for shRNAs (b-e, and g-h) or transfected with siRNA (f) targeting indicated mRNA transcripts, or a non-targeting (NT) shRNA or siRNA. b-d, 96 h post transduction, mRNA levels, protein levels and cell viability were evaluated. b, ATLs mRNA transcript levels were evaluated by RT-qPCR and values corrected using HPRT. Graphs show average percent change compared to NT shRNA for 3 independent experiments. c, ATL protein levels evaluated by western blot. Graph shows average protein levels compared to NT shRNA treated cells from 3 experimental replicates. d, Graph shows mean cell viability as percent survival compared to NT shRNA treated cells. e, 48 h after transduction cells were infected with DV (MOI=1) for 48 h. DV titre were determined by PFU assay. Graph show average fold change in PFU/mL (titre). * and ** - p values lower than 0.05 or 0.01, respectively, determined using one-way ANOVA with a Dunnett’s multiple comparison analysis. RTN, reticulon; LNP, lunapark. f, 96 h post transduction, cells were lysed for RNA analysis. Graph shows average percent change vs. NTshRNA for 3 independent experiments g-h, 48 h after transduction cells were infected with DV or RVFV (MOI=1) for 48 h. DV titre were evaluated using a PFU assay and RVFV replication was determined by luciferase assay. Graphs show average fold change in PFU/mL (titre) or average fold change in RLU/mL (replication) relative to NT shRNA expressing cells for three independent experiments. For all graphs, error bars show SEM and N= ≥ 3 biological replicates.

Extended Data Fig. 2 Virus protein localization in ATL depleted cells.

48 h after lentiviral transduction of shRNA expression constructs, cells were infected with DV for 48 h. Cells were fixed with paraformaldehyde and viewed by immunofluorescence microscopy using LipidTOX or antibodies directed against capsid, Env, NS4B, NS3, Climp63 or RTN3. Scale bars, 10 µm, inset 5 µm.

Extended Data Fig. 3 Effects of ATL depletion on virus-induced membrane alterations.

Cells were transduced with constructs encoding for shRNAs directed against ATL2 or ATL3, or a NT shRNA. a, 48 h after transduction, cells were infected with DV and 48 h later fixed and processed for viewing by TEM. Invaginated vesicles (yellow dots) and viral particles (blue dots) were determined by counting using Fiji software. Inserts display magnified views of the boxed area (scale bars, 2 µm; inset, 200 nm). Note the accumulation of ER tubular networks at the cell periphery in ATL3 depleted cells. b-f, Huh7-Lunet cells stably expressing the bacterial T7 RNA polymerase were transduced for 48 h, followed by transfection with a construct encoding for the viral polyprotein and containing the 3’ untranslated region (UTR) of the DV genome (panel b). c, 24 h post transfection, cells were fixed and stained with antibodies specific to the viral NS3 protein and visualized by confocal light microscopy. Scale bars, 10 µm. d, Graph shows the average relative fluorescence signal of NS3 in shRNA transduced cells. N=7 independent samples. e, Representative EM images of polyprotein expressing cells from a total of 3 independent experiments. Scale bars, 200nm. f, VPs in each cell were counted and mean values are represented in the graph (N=23 cells, error bars represent SEM). For all graphs * and ** represent p values lower than 0.05 or 0.01, respectively as determined by 2-tailed T- test.

Extended Data Fig. 4 ATLs associate with distinct DV proteins.

a-c, A549 cells stably expressing HA-ATL2 or HA-ATL3 were infected with DV for 48 h. Cells were then fixed and stained with antibodies directed against NS3 or Env and the HA epitope, respectively. Scale bars, 10 µm. d, Pearson’s colocalization coefficients were calculated for cells from panels a-c. The graph shows the average value from 20 cells for each condition. Error bars, SEM. e, A549 cells stably expressing HA-ATL2, HA-ATL3, HA-CANX, or an empty plasmid were infected with DV for 48 h. Cells were lysed and HA-tagged proteins were immunoprecipitated with anti-HA beads. Inputs and precipitated proteins were analysed by western blot using NS2B- and NS4B- specific antibodies. Breaks between adjacent blots indicate lanes not relevant to the experiment were removed. f-h, A549 cells stably expressing HA-ATL3 were infected with DV for 48 h. Cells were fixed and stained with antibodies directed against virus proteins or dsRNA (RED) and the HA epitope (green). Pearson’s colocalization coefficients for merge images are given in the top right corners. Images were taken using an Abberior instruments STED microscope. Scale bars, confocal 10 µm, STED 1 µm, inset 100 nm. i, Average Pearson’s colocalization coefficients were calculated for the fluorescent signal corresponding to the HA-tagged ATL3 compared to those from the indicated viral proteins in STED microscopy images. Graph shows the average Pearson’s colocalization coefficients calculated for 10 cells. Error bars, SEM.

Source data

Extended Data Fig. 5 Production and testing of endogenously tagged ATL3.

a, Schematic representation of the cloning strategy used for endogenous tagging of ATL3. NVD, N-terminal variable domain; 3HB, three-bundle Helix; TM, transmembrane region; CTA, C- terminal amphipathic-helix domain. The tagging cassette includes the 11th beta-strand of GFP (GFP11) and the FLAG-tag. b, Individual ATL3-ET (Endogenously Tagged) cell clones were lysed and lysates were analysed by western blot using the indicated antibodies. N=2 biological replicates. c, Cells expressing the endogenously tagged ATL3 were fixed and stained for FLAG or the ER marker PDI. Fluorescence signals specific for antibodies or GFP were visualized by confocal microscopy. Scale bar, 10 µm. d-f, A549 cells expressing endogenously-tagged ATL3 were infected with DV for 48 h. d, Cells were fixed and stained with NS3 (red) or FLAG-specific antibodies (green). Scale bars, 10 µm. e, Cells were lysed and FLAG-tagged proteins were precipitated with anti-FLAG magnetic beads. Inputs and captured protein complexes were evaluated by western blot using antibodies of given specificities. Actin served as loading and specificity control. Breaks between adjacent blots indicate lanes not relevant to the experiment were removed. N=2 biological replicates. f, Titre of infectious extracellular DV were determined by PFU assay. Graph shows the average fold change in PFU ml−1 compared to wild type cells for CRISPR–Cas9 control (Ctrl) or ATL3-ET cells over 3 biological replicates. Error bars, SEM.

Source data

Extended Data Fig. 6 Production of ATL KO cells and effects of ATL mutations on viral replication and subcellular localization.

a-e, A549 cells were transduced with vectors expressing CRISPR–Cas9 as well as a guide RNA directed towards ATL1, ATL2, ATL3, or a non-target guide RNA (Ctrl). a, Knock out or control cells were lysed and protein levels were determined by western blot using given antibodies. GapDH served as loading control. b, Knock out or control cell pools were infected with ZV or DV for 48 h followed by quantification of extracellular virus titre. Graphs show average fold change in PFU ml−1 for each cell line compared to control cells over 3 independent experiments. ** and *** represent p values lower than 0.01 or 0.001, respectively as determined by 2-tailed T-test. c-e, Knockout or control cell pools were transduced with lentiviruses encoding for the ATL variant given on the bottom of each. c-d, 72 h after transduction, cell viability was determined using celltiter glow measuring intracellular ATP levels. Graphs show the average fold change in cell viability compared to control A549 cells. Lower dashed line shows the cut off of 80% viability. N=3 biological replicates. e, 24 h after transduction, cells were infected with DV for 48 h followed by evaluation of virus production using PFU assay. Graph shows the average PFU ml−1 as fold change compared to ctrl cells for 3 independent experiments. Lower dashed line indicates the difference between ATL3 KO cells and ctrl cells, both transduced with an empty plasmid. For all graphs, error bars show SEM. f-g, A549 cells were transduced with constructs encoding for the indicated ATL variants. 72 h after transduction, cells were fixed and stained with antibodies directed against the HA epitope (green) or the ER marker reticulon 3 (RTN3; red). Scale bars, 10 µm.

Source data

Extended Data Fig. 7 ATL2 and ATL3 overexpression does not alter the cellular proteome.

Proteomic analysis of A549 cells stably expressing HA-ATL2, HA-ATL3, HA-CANX, or an empty plasmid. a, Heat map of log2-transformed LFQ intensities for each individual replicate in rainbow colours (see colour scale). b-c, Volcano plots of the p values vs. the log2 protein abundance differences between HA-tagged ATL2- and ATL3-overexpressing cells compared to HA-Calnexin (CANX) overexpressig cells, with proteins outside the significance lines highlighted (unadjusted two-sided t-test. Permutation based FDR < 0.05, S0 = 1, p<0.05). N=4 independent experiments. For Raw data see Source Data Table 1.

Source data.

Extended Data Fig. 8 ATL interactome and shRNA screen.

a-b, The scatter plot displays ATL2 (a) or ATL3 (b) specific interactors (compared to calnexin (CANX)) in both infected and uninfected cells. Schematics of the variables compared are shown in the bottom right of each scatter plot. Significantly enriched or depleted proteins are shown in red (N = 4 independent experiments. Welch’s T-test unadjusted two-sided P ≤ 0.05; |log2(fold-change)| ≥ 1). c, Heat map showing imputed log2- transformed iBAQ intensities for each individual replicate in rainbow colours (see colour scale). Only the bait proteins and the selected cellular interaction partners used for the RNAi screen are depicted in the plot. d, Knockout or control cells were transduced with lentivirus encoding for shRNAs (3/gene) targeting the genes specified in the left of the panel. 72 h after transduction, cell viability was tested. Graphs show the average change in cell viability, as determined by intracellular ATP quantification, for each treatment compared to control cells that were transduced with the non-target shRNA vectors. N=3 independent experiments. Error bars, SEM e, A549 cells stably expressing HA-ATL2, HA-ATL3, HA-CANX (calnexin) or transduced with the empty vector were infected with DV for 48 h. Cells were lysed and HA-tagged proteins were captured with anti-HA beads. Inputs and precipitated protein were determined by western blot and probing for the indicated proteins. Red numbers below the ARF4 panel indicate efficiency of ARF4 pulldown compared to bait protein over an average of 3 experiments. Breaks between adjacent blots indicate non-relevant lanes were removed.

Source data

Extended Data Fig. 9 Effects of ATL3 depletion on virus particle maturation and the secretory pathway.

a, Cells were transduced with lentiviruses encoding for ARF4, ARF5 or non-targeting (NT) shRNAs. 72 h later RNA levels were quantified by RT-qPCR. Shown is the average fold change in viral RNA levels, corrected for HRPT. b-c, Cells were transduced with lentiviruses encoding for ATL2, ATL3 or NT shRNAs. b, After 72 h cells were transfected with a construct encoding for Gaussia luciferase and luciferase secretion was measured over 10 h. Graph shows the average levels of secreted luciferase compared to the 0 h time point. c, 72 h post transduction, cells were transfected with VSV-G_ts045_GFP and 8 h later incubated at 40 °C. 16 h later temperature was lowered to 32 °C and cells were imaged by confocal microscopy. Graph shows the means and SEM of perinuclear fluorescence intensity distribution for each condition. d-h, A549 cells were transduced with constructs of given specificities and cultured for 48 h. d, Cells were infected with ZV, and 48 h later viral RNA levels were determined by RT-qPCR (left panel). Intracellular viral RNA levels were corrected for HRPT. Titres of infectious virus contained in cell lysates and culture supernatants were determined using a PFU assay (right panel). For both panels, average fold changes are shown. e, Levels of prM and NS1 released from DV infected cells were calculated by quantifying western blots using Fiji software. Values were normalized to NT shRNA transduced cells (horizontal line). f, Cells were infected with DV for 48 h. RNA levels were determined by RT-qPCR; graph shows average fold change. g-h, Extracellular proteins were evaluated using western blot. h, Levels of prM released from DV infected cells were calculated using Fiji software. Values are displayed relative to NT shRNA transduced cells. All graphs show means and SEM derived from an average of 3 independent experiments. Significance was determined relative to NT shRNA transduced cells. * or ** represent p values < 0.05 or 0.01, respectively as determined by one-way ANOVA with a Dunnett’s multiple comparison analysis.

Source data

Extended Data Fig. 10 Effects of ATL3 depletion on specific host protein localization.

A549 cells were transduced with constructs encoding for shRNA directed against the indicated gene, or a NT shRNA. a, 48 h post transduction cells were infected with DV for 48 h. Cells were then fixed and the indicated proteins or structures were visualized by immune staining and confocal microscopy. b-e, 96 h after transduction, cells were fixed and stained with the antibodies of given specificity. After incubation with secondary, antibody fluorescence signal was visualized by confocal microscopy. f, shRNA transduced cells expressing the furin reporter protein CD4-Fu were fixed 72 h after transduction. The subcellular localization of the furin reporter was evaluated using immunofluorescence staining and confocal microscopy. Nuclear DNA was stained with DAPI. All scale bars, 10 µm. g, Transduced cells expressing the furin reporter protein were fixed 72 h after transduction and stained with anti-CD4 antibodies. The average total fluorescence levels of CD4-Fu were determined for ≥50 ctrl or ATL3 KD cells. Error bars, SEM.

Supplementary information

Supplementary Information

Supplementary Tables 1-7

Reporting Summary

Supplementary Table 8

Atlastin interactome Mass Spectrometry data

Supplementary Video

Furin internalization. A549 cells stably expressing the furin reporter (CD4-Fu) were transduced with constructs encoding for shRNA directed against the indicated gene or a non-targeting (NT) shRNA. 72 h post transduction, cells were imaged at 1 minute intervals. Following the 2 min time point, anti-CD4 fluor-conjugated antibodies were added and imaging continued for a total of 18 min. Arrows denote foci of intracellular CD4-furin accumulation. The video shows a representative cell from 3 biologically independent replicates.

Source data

Source Data Fig. 2

Unprocessed western blots

Source Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 6

Unprocessed western blots

Source Data Extended Data Fig. 7

Raw data for proteomics analysis

Source Data Extended Data Fig. 8

Unprocessed western blots

Source Data Extended Data Fig. 9

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Neufeldt, C.J., Cortese, M., Scaturro, P. et al. ER-shaping atlastin proteins act as central hubs to promote flavivirus replication and virion assembly. Nat Microbiol 4, 2416–2429 (2019). https://doi.org/10.1038/s41564-019-0586-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0586-3

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