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:

Herpesviruses induce aggregation and selective autophagy of host signalling proteins NEMO and RIPK1 as an immune-evasion mechanism

Abstract

Viruses manipulate cellular signalling by inducing the degradation of crucial signal transducers, usually via the ubiquitin–proteasome pathway. Here, we show that the murine cytomegalovirus (Murid herpesvirus 1) M45 protein induces the degradation of two cellular signalling proteins, the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) essential modulator (NEMO) and the receptor-interacting protein kinase 1 (RIPK1), via a different mechanism: it induces their sequestration as insoluble protein aggregates and subsequently facilitates their degradation by autophagy. Aggregation of target proteins requires a distinct sequence motif in M45, which we termed ‘induced protein aggregation motif’. In a second step, M45 recruits the retromer component vacuolar protein sorting 26B (VPS26B) and the microtubule-associated protein light chain 3 (LC3)-interacting adaptor protein TBC1D5 to facilitate degradation of aggregates by selective autophagy. The induced protein aggregation motif is conserved in M45-homologous proteins of several human herpesviruses, including herpes simplex virus, Epstein–Barr virus and Kaposi’s sarcoma-associated herpesvirus, but is only partially conserved in the human cytomegalovirus UL45 protein. We further show that the HSV-1 ICP6 protein induces RIPK1 aggregation and degradation in a similar fashion to M45. These data suggest that induced protein aggregation combined with selective autophagy of aggregates (aggrephagy) represents a conserved viral immune-evasion mechanism.

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: MCMV M45 induces protein aggregation.
Fig. 2: A short sequence within M45 is required for aggregate formation.
Fig. 3: The IPAM is required for M45 interactions, inhibition of necroptosis and viral dissemination in vivo.
Fig. 4: M45 recruits VPS26B and TBC1D5 to promote degradation of NEMO.
Fig. 5: Induced protein aggregation and degradation by HSV-1 ICP6.
Fig. 6: Graphical summary.

Similar content being viewed by others

Data availability

The datasets generated and analysed during the course of this study are available from the corresponding author upon request without restrictions. Uncropped western blot images of all figures in the manuscript (Figs. 15, Extended Data Figs. 3, 4, 6 and 7) and numerical data with statistical analysis (Figs. 2, 3 and 5) are provided as supplementary source data.

References

  1. Takeuchi, O. & Akira, S. Pattern recognition receptors and inflammation. Cell 140, 805–820 (2010).

    CAS  PubMed  Google Scholar 

  2. Orzalli, M. H. & Kagan, J. C. Apoptosis and necroptosis as host defense strategies to prevent viral infection. Trends Cell Biol. 27, 800–809 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Boutell, C. & Everett, R. D. Regulation of alphaherpesvirus infections by the ICP0 family of proteins. J. Gen. Virol. 94, 465–481 (2013).

    CAS  PubMed  Google Scholar 

  4. Lanfranca, M. P., Mostafa, H. H. & Davido, D. J. HSV-1 ICP0: an E3 ubiquitin ligase that counteracts host intrinsic and innate immunity. Cells 3, 438–454 (2014).

    PubMed  PubMed Central  Google Scholar 

  5. Mizushima, N., Yoshimori, T. & Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu. Rev. Cell. Dev. Biol. 27, 107–132 (2011).

    CAS  PubMed  Google Scholar 

  6. Birgisdottir, A. B., Lamark, T. & Johansen, T. The LIR motif—crucial for selective autophagy. J. Cell Sci. 126, 3237–3247 (2013).

    CAS  PubMed  Google Scholar 

  7. Zaffagnini, G. & Martens, S. Mechanisms of selective autophagy. J. Mol. Biol. 428, 1714–1724 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Choi, Y., Bowman, J. W. & Jung, J. U. Autophagy during viral infection—a double-edged sword. Nat. Rev. Microbiol. 16, 341–354 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kudchodkar, S. B. & Levine, B. Viruses and autophagy. Rev. Med. Virol. 19, 359–378 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Dong, X. & Levine, B. Autophagy and viruses: adversaries or allies? J. Innate Immun. 5, 480–493 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Pellet, P. E. & Roizman, B. in Fields Virology 6th edn (eds Knipe, D., M. & Howley, P. M.) 1802–1822 (Lippincott Williams & Wilkins, 2013).

  12. Brune, W., Ménard, C., Heesemann, J. & Koszinowski, U. H. A ribonucleotide reductase homolog of cytomegalovirus and endothelial cell tropism. Science 291, 303–305 (2001).

    CAS  PubMed  Google Scholar 

  13. Mack, C., Sickmann, A., Lembo, D. & Brune, W. Inhibition of proinflammatory and innate immune signaling pathways by a cytomegalovirus RIP1-interacting protein. Proc. Natl Acad. Sci. USA 105, 3094–3099 (2008).

    CAS  PubMed  Google Scholar 

  14. Upton, J. W., Kaiser, W. J. & Mocarski, E. S. Cytomegalovirus M45 cell death suppression requires receptor-interacting protein (RIP) homotypic interaction motif (RHIM)-dependent interaction with RIP1. J. Biol. Chem. 283, 16966–16970 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Upton, J. W., Kaiser, W. J. & Mocarski, E. S. Virus inhibition of RIP3-dependent necrosis. Cell Host Microbe 7, 302–313 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Rebsamen, M. et al. DAI/ZBP1 recruits RIP1 and RIP3 through RIP homotypic interaction motifs to activate NF-κB. EMBO Rep. 10, 916–922 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Upton, J. W., Kaiser, W. J. & Mocarski, E. S. DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe 11, 290–297 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Maelfait, J. et al. Sensing of viral and endogenous RNA by ZBP1/DAI induces necroptosis. EMBO J. 36, 2529–2543 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Fliss, P. M. et al. Viral mediated redirection of NEMO/IKKγ to autophagosomes curtails the inflammatory cascade. PLoS Pathog. 8, e1002517 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Krause, E., de Graaf, M., Fliss, P. M., Dölken, L. & Brune, W. Murine cytomegalovirus virion-associated protein M45 mediates rapid NF-κB activation after infection. J. Virol. 88, 9963–9975 (2014).

    PubMed  PubMed Central  Google Scholar 

  21. Hüttmann, J., Krause, E., Schommartz, T. & Brune, W. Functional comparison of molluscum contagiosum virus vFLIP MC159 with murine cytomegalovirus M36/vICA and M45/vIRA proteins. J. Virol. 90, 2895–2905 (2015).

    PubMed  Google Scholar 

  22. Carisey, A., Stroud, M., Tsang, R. & Ballestrem, C. Fluorescence recovery after photobleaching. Methods Mol. Biol. 769, 387–402 (2011).

    CAS  PubMed  Google Scholar 

  23. Brangwynne, C. P., Mitchison, T. J. & Hyman, A. A. Active liquid-like behavior of nucleoli determines their size and shape in Xenopus laevis oocytes. Proc. Natl Acad. Sci. USA 108, 4334–4339 (2011).

    CAS  PubMed  Google Scholar 

  24. Link, C. D. et al. Conversion of green fluorescent protein into a toxic, aggregation-prone protein by C-terminal addition of a short peptide. J. Biol. Chem. 281, 1808–1816 (2006).

    CAS  PubMed  Google Scholar 

  25. Huang, Z. et al. RIP1/RIP3 binding to HSV-1 ICP6 initiates necroptosis to restrict virus propagation in mice. Cell Host Microbe 17, 229–242 (2015).

    CAS  PubMed  Google Scholar 

  26. Lembo, D. et al. The ribonucleotide reductase R1 homolog of murine cytomegalovirus is not a functional enzyme subunit but is required for pathogenesis. J. Virol. 78, 4278–4288 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Mandal, P. et al. RIP3 induces apoptosis independent of pronecrotic kinase activity. Mol. Cell 56, 481–495 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Seaman, M. N., McCaffery, J. M. & Emr, S. D. A membrane coat complex essential for endosome-to-Golgi retrograde transport in yeast. J. Cell. Biol. 142, 665–681 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Swarbrick, J. D. et al. VPS29 is not an active metallo-phosphatase but is a rigid scaffold required for retromer interaction with accessory proteins. PLoS ONE 6, e20420 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Seaman, M. N. The retromer complex—endosomal protein recycling and beyond. J. Cell Sci. 125, 4693–470 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Collins, B. M. et al. Structure of Vps26B and mapping of its interaction with the retromer protein complex. Traffic 9, 366–379 (2008).

    CAS  PubMed  Google Scholar 

  32. Bugarcic, A. et al. Vps26A and Vps26B subunits define distinct retromer complexes. Traffic 12, 1759–1773 (2011).

    CAS  PubMed  Google Scholar 

  33. Popovic, D. et al. Rab GTPase-activating proteins in autophagy: regulation of endocytic and autophagy pathways by direct binding to human ATG8 modifiers. Mol. Cell Biol. 32, 1733–1744 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Johansen, T. & Lamark, T. Selective autophagy mediated by autophagic adapter proteins. Autophagy 7, 279–296 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Popovic, D. & Dikic, I. TBC1D5 and the AP2 complex regulate ATG9 trafficking and initiation of autophagy. EMBO Rep. 15, 392–401 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Lembo, D. & Brune, W. Tinkering with a viral ribonucleotide reductase. Trends Biochem. Sci. 34, 25–32 (2009).

    CAS  PubMed  Google Scholar 

  37. Guo, H. et al. Herpes simplex virus suppresses necroptosis in human cells. Cell Host Microbe 17, 243–251 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Yu, X. et al. Herpes simplex virus 1 (HSV-1) and HSV-2 mediate species-specific modulations of programmed necrosis through the viral ribonucleotide reductase large subunit R1. J. Virol. 90, 1088–1095 (2016).

    CAS  PubMed  Google Scholar 

  39. Alexander, D. E., Ward, S. L., Mizushima, N., Levine, B. & Leib, D. A. Analysis of the role of autophagy in replication of herpes simplex virus in cell culture. J. Virol. 81, 12128–12134 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lussignol, M. et al. The herpes simplex virus 1 Us11 protein inhibits autophagy through its interaction with the protein kinase PKR. J. Virol. 87, 859–871 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Lamark, T. & Johansen, T. Aggrephagy: selective disposal of protein aggregates by macroautophagy. Int. J. Cell Biol. 2012, 736905 (2012).

    PubMed  PubMed Central  Google Scholar 

  42. Li, J. et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150, 339–350 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ali, M., Roback, L. & Mocarski, E. S. Herpes simplex virus 1 ICP6 impedes TNF receptor 1-induced necrosome assembly during compartmentalization to detergent-resistant membrane vesicles. J. Biol. Chem. 294, 991–1004 (2019).

    CAS  PubMed  Google Scholar 

  44. Cheng, A. Z. et al. Epstein–Barr virus BORF2 inhibits cellular APOBEC3B to preserve viral genome integrity. Nat. Microbiol. 4, 78–88 (2019).

    CAS  PubMed  Google Scholar 

  45. Dufour, F. et al. The ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 protect cells against TNF-α and FasL-induced apoptosis by interacting with caspase-8. Apoptosis 16, 256–271 (2011).

    CAS  PubMed  Google Scholar 

  46. Kwon, K. M., Oh, S. E., Kim, Y. E., Han, T. H. & Ahn, J. H. Cooperative inhibition of RIP1-mediated NF-κB signaling by cytomegalovirus-encoded deubiquitinase and inactive homolog of cellular ribonucleotide reductase large subunit. PLoS Pathog. 13, e1006423 (2017).

    PubMed  PubMed Central  Google Scholar 

  47. Carra, S., Seguin, S. J. & Landry, J. HspB8 and Bag3: a new chaperone complex targeting misfolded proteins to macroautophagy. Autophagy 4, 237–239 (2008).

    CAS  PubMed  Google Scholar 

  48. Meriin, A. B. et al. Hsp70–Bag3 complex is a hub for proteotoxicity-induced signaling that controls protein aggregation. Proc. Natl Acad. Sci. USA 115, E7043–E7052 (2018).

    CAS  PubMed  Google Scholar 

  49. Paul, P. & Münz, C. Autophagy and mammalian viruses: roles in immune response, viral replication, and beyond. Adv. Virus Res. 95, 149–195 (2016).

    CAS  PubMed  Google Scholar 

  50. Kim, E. et al. Implication of mouse Vps26b–Vps29–Vps35 retromer complex in sortilin trafficking. Biochem. Biophys. Res. Commun. 403, 167–171 (2010).

    CAS  PubMed  Google Scholar 

  51. Bresnahan, W. A., Hultman, G. E. & Shenk, T. Replication of wild-type and mutant human cytomegalovirus in life-extended human diploid fibroblasts. J. Virol. 74, 10816–10818 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Jordan, S. et al. Virus progeny of murine cytomegalovirus bacterial artificial chromosome pSM3fr show reduced growth in salivary glands due to a fixed mutation of MCK-2. J. Virol. 85, 10346–10353 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Tischer, B. K., Smith, G. A. & Osterrieder, N. En passant mutagenesis: a two step markerless red recombination system. Methods Mol. Biol. 634, 421–430 (2010).

    CAS  PubMed  Google Scholar 

  54. Arase, H., Mocarski, E. S., Campbell, A. E., Hill, A. B. & Lanier, L. L. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 296, 1323–1326 (2002).

    CAS  PubMed  Google Scholar 

  55. Handke, W. et al. Viral inhibition of BAK promotes murine cytomegalovirus dissemination to salivary glands. J. Virol. 87, 3592–3596 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Brune, W., Hengel, H. & Koszinowski, U. H. A mouse model for cytomegalovirus infection. Curr. Protoc. Immunol. 43, 19.7.1–19.7.13 (2001).

    Google Scholar 

  57. Mahy, B. W. J. & Kangro, H. O. Virology Methods Manual (Academic Press, 1996).

  58. Tanaka, M., Kagawa, H., Yamanashi, Y., Sata, T. & Kawaguchi, Y. Construction of an excisable bacterial artificial chromosome containing a full-length infectious clone of herpes simplex virus type 1: viruses reconstituted from the clone exhibit wild-type properties in vitro and in vivo. J. Virol. 77, 1382–1391 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Ashida, H. et al. A bacterial E3 ubiquitin ligase IpaH9.8 targets NEMO/IKKγ to dampen the host NF-κB-mediated inflammatory response. Nat. Cell Biol. 12, 66–73 (2010).

    CAS  PubMed  Google Scholar 

  60. Ostermann, E. et al. Activation of E2F-dependent transcription by the mouse cytomegalovirus M117 protein affects the viral host range. PLoS Pathog. 14, e1007481 (2018).

    PubMed  PubMed Central  Google Scholar 

  61. van de Weijer, M. L. et al. A high-coverage shRNA screen identifies TMEM129 as an E3 ligase involved in ER-associated protein degradation. Nat. Commun. 5, 3832 (2014).

    PubMed  PubMed Central  Google Scholar 

  62. Chen, D. & Huang, S. Nucleolar components involved in ribosome biogenesis cycle between the nucleolus and nucleoplasm in interphase cells. J. Cell Biol. 153, 169–176 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Montespan, C. et al. Multi-layered control of Galectin-8 mediated autophagy during adenovirus cell entry through a conserved PPxY motif in the viral capsid. PLoS Pathog. 13, e1006217 (2017).

    PubMed  PubMed Central  Google Scholar 

  64. Corpet, F. Multiple sequence alignment with hierarchical clustering. Nucleic Acids Res. 16, 10881–10890 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Robert, X. & Gouet, P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res. 42, W320–W324 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Connor, J. Bertin and E. Mocarski for RIPK3 kinase-dead mice, S. Jonjic, Y. Kawaguchi and R. Teasdale for reagents, F. Giraudo for technical assistance and T. Potgieter for critical readings of the manuscript. This study was supported by funding from Deutsche Forschungsgemeinschaft (BR 1730/3-2 to WB). The Heinrich Pette Institute is supported by the Free and Hanseatic City of Hamburg and the Federal Ministry of Health. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

E.M. designed, performed and analysed most experiments. R.S. performed a few biochemical experiments. E.K. and S.L. performed SILAC and AP–MS analyses. A.S. supervised the mass spectrometry analyses. E.C. performed and analysed FRAP experiments. M.R. generated and analysed TBC1D5-deficient cell clones. C.S. and R.R. performed correlative light and electron microscopy analyses. Y.-H.K. provided mice and biochemical reagents. V.J.L. provided biochemical reagents. E.O. performed in vivo experiments. W.B. designed and supervised the study, acquired funding and provided resources. E.M. and W.B. wrote and edited the manuscript.

Corresponding author

Correspondence to Wolfram Brune.

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 M45 induced aggregates.

(a) Atg5-/- MEFs were transfected with an M45-mCherry plasmid. 24 h post transfection cells were fixed and analyzed by fluorescence microscopy. Nuclei were stained with Hoechst 33342. Scale bar, 10 µm. (b) NIH-3T3 cells transfected with plasmids expressing HA-tagged full-length M45 or M45-Ct3. HA-tagged proteins were detected by immunofluorescence (green), protein aggregates by using the ProteoStat dye (red). (c) Another two ultrathin sections of the same WT MEF cell shown in Fig. 1f (ix – xi). The arrow points to a different aggregate than the one in Fig. 1f. Magnification of the same aggregate (A) close to an autophagosome (*) in the two different sections (x – xii). Scale bars, 5 µm (ix, xi) and 200 nm (x, xii). (d) Maximum intensity projection of NIH-3T3 cell transfected with a plasmid encoding M45-mCherry and observed 6 h post transfection by live cell imaging for 30 h. Scale bar, 10 µm. These data are representative of three (a-b) or two (d) biologically independent experiments. This data (c) is representative of three independent infected cells per group where at least 10 different sections (50 nm) per cells were analyzed.

Extended Data Fig. 2 Proteins co-purifying with M45 detected by AP-MS.

NIH-3T3 cells were labelled by SILAC and infected with MCMV WT or MCMV-M45HA. Cell lysates were harvested 15 hpi and subjected to anti-HA affinity-purification. Purified were analyzed by mass spectrometry.

Extended Data Fig. 3 M45 interacts with VPS26B, independent of VPS26B’s interaction with VPS35, and TBC1D5.

(a) Schematic of WT and mutant VPS26B. (b) HEK-293A cells were co-transfected with VPS26B-myc and VPS35-Flag plasmids. Immunoprecipitations were done as indicated. (c) HEK-293A cells co-transfected with plasmids expressing M45- HA and myc-tagged VPS26B or VPS26A (negative control). M45-HA was immunoprecipitated. (d) NIH-3T3-Vps26B-myc cells infected with MCMV-M45HA, MCMV-M45mut2HA or HA- tagged M45 C-terminal truncation mutants (MOI 3). Immunoprecipitation was done with an anti-HA antibody. (e) MEFs were infected with MCMV-M45HA or MCMVΔM45 (MOI 3). (f) VPS35, VPS26A, VPS29, and viral protein levels were determined by immunoblot at different times post infection. HEK-293A cells co-transfected with plasmids expressing M45-HA and Flag-TBC1D5 or Flag-IFI16 (negative control) or Flag-NEMO (positive control). HA was immunoprecipitated. (g) HEK-293A cells co-transfected with plasmids expressing M45- HA full-length or C-terminus truncation mutants (HA tagged) and Flag-TBC1D5. Immunoprecipitation was done with an anti-HA antibody. (h) WT and Vps26b-/- MEFs were infected with MCMV-M45HA (MOI 3). Immunoprecipitation was done with an anti-HA antibody. Immunoblot labels are in kDa. These data are representative of two (b-e-h) or three (c-d-f-g) biologically independent experiments.

Source data

Extended Data Fig. 4 HSV-1 ICP6 induces aggregate formation.

(a) Ripk3-/- fibroblasts were transfected with plasmids expressing WT or mutant ICP6. 24 h post transfection protein aggregates were detected by using the ProteoStat dye (red) and HA-tagged ICP6 by immunofluorescence (green). Scale bar, 10 µm. (b) HFF were infected with WT or ICP6mut HSV-1 (MOI 1). 24 hpi cells were fixed and stained for LC3BII (green). Nuclei were stained with Hoechst 33342. Scale bar, 10 µm. (c) HFF infected as in b. LC3BII was detected by immunoblot at 8 and 24 hpi. (d) HFF were infected with HSV-1 ICP6HA or ICP6mutHA (MOI 1). 24 hpi cells were fixed and stained for gamma-tubulin (green) and HA (red). Nuclei were stained with Hoechst 33342. Scale bar, 10 µm. Immunoblot labels are in kDa. These data are representative of two (a-b-c) or three (d) biologically independent experiments.

Source data

Extended Data Fig. 5 M45 aggregates co-localize with LC3BII.

(a, b) NIH-3T3 infected with MCMV-M45HA or MCMV- M45mut2HA (MOI 3). 24 hpi cells were fixed and stained for HA (red) and either LC3BII (a) or Caveolin-1 (b) (green). Nuclei were stained with Hoechst 33342. Scale bar, 10 µm. These data (a-b) are representative of three biologically independent experiments.

Extended Data Fig. 6 M45-interacting regions in NEMO and RIPK1 do not share a common motif.

(a) HEK-293A cells were co-transfected with plasmids expressing M45-HA and Flag-tagged RIPK1. Full-length (FL) and deletion mutants lacking the N-terminus, the C-terminus, or the death domain (DD) were used. M45-HA was immuno- precipitated and co-precipitating proteins were detected by immunoblot. (b) HEK-293A cells were co-transfected with plasmids expressing M45-HA and Flag-tagged NEMO (FL or N- terminal truncation mutants). M45-HA was immuno- precipitated and co-precipitating proteins were detected by immunoblot. (c) Sequence alignment of M45-interacting regions in NEMO and RIPK1 with APOBEC3B. Immunoblot labels (a-b) are in kDa. These data are representative of two (a) or three (b) biologically independent experiments.

Source data

Extended Data Fig. 7 M45 aggregates do not co-localize with HSP70.

(a) NIH-3T3 were infected with MCMV-M45HA or MCMV- M45mut2HA (MOI 3). 24 hpi cells were fixed and immunostained for HA (red) and HSP70 (green). Nuclei were stained with Hoechst 33342. Scale bar, 10 µm. (b) Immunoblot analysis of the soluble and insoluble fractions of MCMV- M45HA and MCMV-M45mut2 infected Atg5-/- MEFs (MOI 5). These data (a-b) are representative of three biologically independent experiments.

Source data

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blot.

Source Data Fig. 2

Unprocessed western blot.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Unprocessed western blot.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Unprocessed western blot.

Source Data Fig. 5

Unprocessed western blot.

Source Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 3

Unprocessed western blot.

Source Data Extended Data Fig. 4

Unprocessed western blot.

Source Data Extended Data Fig. 6

Unprocessed western blot.

Source Data Extended Data Fig. 7

Unprocessed western blot.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Muscolino, E., Schmitz, R., Loroch, S. et al. Herpesviruses induce aggregation and selective autophagy of host signalling proteins NEMO and RIPK1 as an immune-evasion mechanism. Nat Microbiol 5, 331–342 (2020). https://doi.org/10.1038/s41564-019-0624-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0624-1

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