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:

Norovirus MLKL-like protein initiates cell death to induce viral egress

Abstract

Non-enveloped viruses require cell lysis to release new virions from infected cells, suggesting that these viruses require mechanisms to induce cell death. Noroviruses are one such group of viruses, but there is no known mechanism that causes norovirus infection-triggered cell death and lysis1,2,3. Here we identify a molecular mechanism of norovirus-induced cell death. We found that the norovirus-encoded NTPase NS3 contains an N-terminal four-helix bundle domain homologous to the membrane-disruption domain of the pseudokinase mixed lineage kinase domain-like (MLKL). NS3 has a mitochondrial localization signal and thus induces cell death by targeting mitochondria. Full-length NS3 and an N-terminal fragment of the protein bound the mitochondrial membrane lipid cardiolipin, permeabilized the mitochondrial membrane and induced mitochondrial dysfunction. Both the N-terminal region and the mitochondrial localization motif of NS3 were essential for cell death, viral egress from cells and viral replication in mice. These findings suggest that noroviruses have acquired a host MLKL-like pore-forming domain to facilitate viral egress by inducing mitochondrial dysfunction.

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: Norovirus egress requires NINJ1-mediated plasma membrane rupture.
Fig. 2: The N-terminal domain of norovirus NS3 directly triggers programmed cell death and has homology to the MLKL four-helix bundle domain.
Fig. 3: Norovirus NS3 permeabilizes the mitochondrial membrane and induces mitochondrial disfunction.
Fig. 4: NS3-triggered cell death is essential for norovirus egress.

Similar content being viewed by others

Data availability

All data are available in the main paper, supplementary information and via figshare https://doi.org/10.6084/m9.figshare.21586098. All reagents are available from the authors under a material transfer agreement with University of Texas Southwestern Medical Center. Source data are provided with this paper.

References

  1. Karst, S. M., Wobus, C. E., Goodfellow, I. G., Green, K. Y. & Virgin, H. W. Advances in norovirus biology. Cell Host Microbe 15, 668–680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Glass, R. I., Parashar, U. D. & Estes, M. K. Norovirus gastroenteritis. N. Engl. J. Med. 361, 1776–1785 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Karst, S. M. & Tibbetts, S. A. Recent advances in understanding norovirus pathogenesis: norovirus pathogenesis. J. Med. Virol. 88, 1837–1843 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Elde, N. C. & Malik, H. S. The evolutionary conundrum of pathogen mimicry. Nat. Rev. Microbiol. 7, 787–797 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Jorgensen, I., Rayamajhi, M. & Miao, E. A. Programmed cell death as a defence against infection. Nat. Rev. Immunol. 17, 151–164 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jorgensen, I. & Miao, E. A. Pyroptotic cell death defends against intracellular pathogens. Immunol. Rev. 265, 130–142 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. McFadden, N. et al. Norovirus regulation of the innate immune response and apoptosis occurs via the product of the alternative open reading frame 4. PLoS Pathog. 7, e1002413 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Furman, L. M. et al. Cysteine protease activation and apoptosis in murine norovirus infection. Virol. J. 6, 139–139 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Bok, K., Prikhodko, V. G., Green, K. Y. & Sosnovtsev, S. V. Apoptosis in murine norovirus-infected RAW264.7 cells is associated with downregulation of survivin. J. Virol. 83, 3647–3656 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dubois, H. et al. Nlrp3 inflammasome activation and gasdermin D-driven pyroptosis are immunopathogenic upon gastrointestinal norovirus infection. PLoS Pathog. 15, e1007709 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Santiana, M. et al. Vesicle-cloaked virus clusters are optimal units for inter-organismal viral transmission. Cell Host Microbe 24, 208–220.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kayagaki, N. et al. NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 591, 131–136 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Wobus, C. E. et al. Replication of norovirus in cell culture reveals a tropism for dendritic cells and macrophages. PLoS Biol. 2, e432 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Degterev, A. et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 1, 112–119 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Hildebrand, J. M. et al. Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death. Proc. Natl Acad. Sci. USA 111, 15072–15077 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Yen, J.-B. et al. Identification and characterization of human norovirus NTPase regions required for lipid droplet localization, cellular apoptosis, and interaction with the viral p22 protein. Microbiol. Spectr. 9, e00422-21 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Yen, J.-B. et al. Subcellular localization and functional characterization of GII.4 norovirus-encoded NTPase. J. Virol. 92, e01824-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Han, K. R. et al. Nucleotide triphosphatase and RNA chaperone activities of murine norovirus NS3. J. Gen. Virol. 99, 1482–1493 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Li, T.-F. et al. Human norovirus NS3 has RNA helicase and chaperoning activities. J. Virol. 92, e01606-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Cotton, B. T. et al. The norovirus NS3 protein is a dynamic lipid- and microtubule-associated protein involved in viral RNA replication. J. Virol. 91, e02138-16 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Thorne, L. G. & Goodfellow, I. G. Norovirus gene expression and replication. J. Gen. Virol. 95, 278–291 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Campillay-Véliz, C. P. et al. Human norovirus proteins: implications in the replicative cycle, pathogenesis, and the host immune response. Front. Immunol. 11, 961 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jones, M. K. et al. Enteric bacteria promote human and mouse norovirus infection of B cells. Science 346, 755–759 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang, H. et al. Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol. Cell 54, 133–146 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Murphy, J. M. et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39, 443–453 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Green, K. Y. et al. A predominant role for Norwalk-like viruses as agents of epidemic gastroenteritis in Maryland nursing homes for the elderly. J. Infect. Dis. 185, 133–146 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Armenteros, J. J. A., Sønderby, C. K., Sønderby, S. K., Nielsen, H. & Winther, O. DeepLoc: prediction of protein subcellular localization using deep learning. Bioinformatics 33, 3387–3395 (2017).

    Article  CAS  Google Scholar 

  29. Chu, C. T. et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat. Cell Biol. 15, 1197–1205 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu, J. et al. Role of phospholipid scramblase 3 in the regulation of tumor necrosis factor-α-induced apoptosis. Biochemistry 47, 4518–4529 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Yunus, M. A., Chung, L. M. W., Chaudhry, Y., Bailey, D. & Goodfellow, I. Development of an optimized RNA-based murine norovirus reverse genetics system. J. Virol. Methods 169, 112–118 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Nice, T. J., Strong, D. W., McCune, B. T., Pohl, C. S. & Virgin, H. W. A single-amino-acid change in murine norovirus NS1/2 is sufficient for colonic tropism and persistence. J. Virol. 87, 327–334 (2012).

    Article  PubMed  Google Scholar 

  33. Karst, S. M., Wobus, C. E., Lay, M., Davidson, J. & Virgin, H. W. IV STAT1-dependent innate immunity to a Norwalk-like virus. Science 299, 1575–1578 (2003).

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Rogers, C. et al. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 10, 1689 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  35. Bock, F. J. & Tait, S. W. G. Mitochondria as multifaceted regulators of cell death. Nat. Rev. Mol. Cell Bio. 21, 85–100 (2020).

    Article  CAS  Google Scholar 

  36. Evavold, C. L. et al. Control of gasdermin D oligomerization and pyroptosis by the Ragulator–Rag–mTORC1 pathway. Cell 184, 4495–4511.e19 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Petrie, E. J. et al. Viral MLKL homologs subvert necroptotic cell death by sequestering cellular RIPK3. Cell Rep. 28, 3309–3319.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Palmer, S., Chappidi, S., Pinkham, C. & Hancks, D. C. Evolutionary profile for (host and viral) MLKL indicates its activities as a battlefront for extensive counteradaptation. Mol. Biol. Evol. 38, msab256 (2021).

    Article  Google Scholar 

  39. Farag, N. S., Breitinger, U., Breitinger, H. G. & Azizi, M. A. E. Viroporins and inflammasomes: key to understand virus-induced inflammation. Int. J. Biochem. Cell Biol. 122, 105738 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nieva, J. L., Madan, V. & Carrasco, L. Viroporins: structure and biological functions. Nat. Rev. Microbiol. 10, 563–574 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mahdi, L. K. et al. Discovery of a family of mixed lineage kinase domain-like proteins in plants and their role in innate immune signaling. Cell Host Microbe 28, 813–824.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Durbin, J. E., Hackenmiller, R., Simon, M. C. & Levy, D. E. Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease. Cell 84, 443–450 (1996).

    Article  CAS  PubMed  Google Scholar 

  43. He, S. et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-α. Cell 137, 1100–1111 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Ward, V. K. et al. Recovery of infectious murine norovirus using Pol II-driven expression of full-length cDNA. Proc. Natl Acad. Sci. USA 104, 11050–11055 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Orchard, R. C. et al. Discovery of a proteinaceous cellular receptor for a norovirus. Science 353, 933–936 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Baert, L. et al. Detection of murine norovirus by using plaque assay, transfection assay, and real-time reverse transcription–PCR before and after heat exposure. Appl. Environ. Microbiol. 74, 543–546 (2008).

    Article  ADS  CAS  PubMed  Google Scholar 

  47. Hwang, S. et al. Murine norovirus: propagation, quantification, and genetic manipulation. Curr. Protoc. Microbiol. 33, 15K.2.1–15K.2.61 (2014).

    Article  PubMed  Google Scholar 

  48. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Vinjé, J. et al. ICTV virus taxonomy profile: Caliciviridae. J. Gen. Virol. 100, 1469–1470 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Z. G. Wang for providing the pLVX-TRE3G vector; J. Rizo-Rey, Z. G. Wang and K. Yang for helpful discussions and technical assistance; and members of the Reese laboratory for technical assistance. T.A.R. is supported by grants from the NIH (R01AI130020-01A1, U19AI142784), CPRIT (RP200118), and the Pew Scholars Program. D.C.H. is funded by a 1R35GM142689-01 and a Recruitment of First-Time, Tenure-Track Faculty from Cancer Prevention & Research Institute of Texas Award (RR 170047).

Author information

Authors and Affiliations

Authors

Contributions

G.W. and T.A.R. conceived the study. G.W. and D.Z. performed experiments and analysed data. D.C.H. performed sequence and phylogenetic analysis. R.C.O. provided reagents and expertise. G.W. and T.A.R. wrote the paper. All authors read and edited the manuscript.

Corresponding authors

Correspondence to Dustin C. Hancks or Tiffany A. Reese.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewer(s) for their contribution to the peer review of this work. Peer review reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Noroviruses infection triggers programmed cell death.

a, Viability of BMDMs and BV2 infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 12 h. b, LDH released from BMDMs and BV2 infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 12 h. c, Representative flow cytometry pseudocolor dot plots of propidium iodide (PI) and annexin V-stained BMDMs infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 12 h. Cells were first gated off forward and side scatter. d, Bright-field images of BMDMs and BV2 infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 12 h. Scale bar, 100 μm. e, Representative IncuCyte images of SytoxGreen-stained BMDMs and BV2 infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 24 h. Scale bar, 400 μm. Data in (a and b) are pooled from three independent experiments with three technical replicates in each experiment. Data are presented as mean of biological replicates ± s.d. Data in (c,d and e) are representative of three independent experiments.

Source Data

Extended Data Fig. 2 Blockade of apoptosis, pyroptosis, or necroptosis does not rescue MNV-induced cell death in in BMDMs and BV2 cells.

a, Representative bright-field images of BMDMs infected with MNoVCR6 or MNoVCW3 at a MOI of 5 with or without 10 μM zVAD and/or 10 μM necrostatin-1 (Nec-1) for 12 h. Scale bar, 100μm. (b to c) Cytotoxicity of BMDMs infected with MNoVCR6 (b) or MNoVCW3 (c) at a MOI of 5 with or without 10 μM zVAD and/or 10 μM Nec-1 for 24 h. d, Representative flow cytometry pseudocolor dot plots of propidium iodide (PI) and annexin V-stained BMDMs are shown. BMDMs infected with MNoVCW3 and MNoVCR6 at a MOI of 5 with or without 10 μM zVAD and/or 10 μM Nec-1 for 12 h. Cells were first gated off forward and side scatter. eh, LDH released from BMDMs (e and f) and BV2 (g and h) infected with MNoVCW3 and MNoVCR6 (MOI=5) with or without 10 μM zVAD, 10 μM Nec-1, either individually or in combination for 24h. i, Cytotoxicity of BV2 cells infected with MNoVCR6 at a MOI of 1 with or without zVAD for 12 h. BV2 cells treated with 0.5 μM Staurosporine (STS) with or without 20 μM zVAD for 6 h as a positive control for apoptosis. j, BV2 cells were primed with 200 ng/mL LPS for 4 h, followed by 10 μM nigericin (Niger) to activate pyroptotic cell death, or they were infected with MNoVCR6, MOI = 1, 12 h. zVAD (20 μM) was added 30 min before nigericin treatment. After 12h treatment or infection, cell viability measured by ATP levels using CellTiter-Glo kit. k, Cytotoxicity of BV2 cells infected with MNoVCR6 at a MOI of 1 with or without 10 μM Nec-1 for 12 h. BV2 cells treated with 20 μM LPS plus 20 μM zVAD to induce necroptosis and treated with or without 30 μM Nec-1 as a control. After 12h treatment or infection, cell viability measured by ATP levels using CellTiter-Glo kit. Data in (a and d) are representative of three independent experiments. Data in (b,c,e,f,g,h,i,j and k) are pooled from three independent experiments with three technical replicates each. Data are presented as mean of biological replicates ± s.d.

Source Data

Extended Data Fig. 3 Pyroptosis, necroptosis and apoptosis are not required for MNoV-induced cell death.

a, WT and Caspase1/11−/− BMDMs cells were treated with vehicle, 10 μM Nec-1 and infected with MNoVCW3 and MNoVCR6 at a MOI of 1 for 24 h. Cell viability measured by ATP levels. b, GSDMD was deleted from BV2 cells by CRISPR genome editing. GSDMD expression levels in WT and four different clones of knockout BV2 cells. Lysates of indicated cells were blotted with anti-GSDMD antibody and anti-actin antibody. c, Comparison of LDH release in WT and Gsdmd−/− cells infected with MNoVCW3 and MNoVCR6 (MOI = 1) for 24 h. d WT and Gsdmd−/− cells infected with MNoVCW3 and MNoVCR6 (MOI = 1) for 24 h. Cell viability as measured by ATP levels. e, WT BMDMs cells infected with MNoVCW3 and MNoVCR6 at a MOI of 1 or treated with 10 μM zVAD plus 100 ng/ml LPS. Lysates of indicated cells were blotted with anti-MLKL antibody, anti-pMLKL antibody and anti-actin antibody. f, Comparison of ATP levels in MNoVCW3 and MNoVCR6 (MOI = 1) infected WT and Ripk3−/− BMDMs with or without 10 μM zVAD for 24 h. g, MLKL was deleted from BV2 cells by CRISPR genome editing. Lysates of wildtype of a knockout clone of MLKL were blotted with anti-MLKL antibody and anti-actin antibody. h, Comparison of ATP levels in MNoVCW3 and MNoVCR6 (MOI=1) infected WT and Mlkl−/− cells with or without 10 μM zVAD for 24 h. i, Bax and Bak were deleted from BV2 cells by CRISPR genome editing and single cell clones were selected. Lysates of WT or knockout cells were blotted with anti-BAK, anti-BAX and anti-actin antibody. j, Gsdmd/Caspase-3 and Gsdmd/Caspase-9 double knockout BV2 cells were made by CRISPR genome editing and single cell clones were selected. Lysates of indicated cells were blotted with indicated antibodies. k, Comparison of ATP levels in MNoVCW3 and MNoVCR6 (MOI = 1) infected WT and Bax−/− / Bak−/− cells for 12 h. l, Comparison of ATP levels in MNoVCW3 and MNoVCR6 (MOI = 1) infected WT, Gsdmd−/− / Caspase-3−/− and Gsdmd−/− / Caspase-9−/− cells for 12 h. m, WT BMDMs cells infected with MNoVCW3 and MNoVCR6 at a MOI of 1 or treated with 10 μg/mL Tunicamycin for 4 h. Lysates of indicated cells were blotted with anti-CHOP antibody, anti-ATF4 antibody, anti-IRE1a antibody and anti-actin antibody. n, WT and Ire1a−/− BV2 cells infected with MNoVCW3 and MNoVCR6 (MOI = 1) for 24. Cell viability as measured by ATP levels. Data in (a,c,d,f,h,k,l and n) are pooled from three independent experiments with three technical replicates each. Data are presented as mean of biological replicates ± s.d. Data in (b,e,g,i,j and m) are representative of two independent experiments.

Source Data

Extended Data Fig. 4 Norovirus NTPase NS3 directly triggers cell death.

a, Schematic model of MNoV NS3. b, Representative bright-field images of HEK293T cells transfected with N-terminal Flag-tagged constructs encoding NS3-FL, NS3-N or NS3-C of MNoVCR6 or MNoVCW3. Scale bar, 100 μm. c, LDH released from HEK293T cells transfected with Full-length Flag-NS3 of MNoVCR6 or MNoVCW3. d, LDH released from HEK293T cells transfected with Full-length, N- or C-terminal Flag-NS3 of MNoVCR6. e, LDH released from HEK293T cells transfected with Full-length, N- or C-terminal Flag-NS3 of MNoVCW3. f, The immunoblot shows expression of HEK293T cells transfected with Full-length, N- or C-terminal of Flag-NS3. g, Immunoblotting assays of norovirus infection-induced GSDMD and CASPASE-3 cleavage in BMDMs. BMDMs infected with MNoVCW3 and MNoVCR6 (MOI = 5) for 12 h. h, Immunoblotting assays of GSDMD and CASPASE-3 cleavage in BV2 cells stably expressing inducible, C-terminal Flag-tagged constructs of NS3-FL, NS3-N or NS3-C of MNoVCR6 in the absence or presence doxycycline for 12 h. Data in (b) are representative of three independent experiments. Data in (c,d and e) are pooled from three independent experiments with three technical replicates each. Data are presented as mean of biological replicates ± s.d. Data in (f,g and h) are representative of two independent experiments.

Source Data

Extended Data Fig. 5 MNoV and HNoV NS3 N-terminal domain directly triggers cell death in multiple cell types.

NS3-FL, NS3-N or NS3-C fused with C-terminal Flag tag were stably expressed in BV2 cells under a tetracycline-inducible promoter. Protein expression was induced with doxycycline treatment. a, Representative bright-field images of BV2 cells stably expressing inducible constructs in the absence or presence doxycycline for 12 h. Scale bar, 100 μm. b, Immunoblot of NS3 with anti-Flag antibody in the absence or presence doxycycline for 12 h. c, Cytotoxicity of BV2 cells expressing inducible constructs after doxycycline addition with or without 20 μM zVAD for 12 h. d, Representative Incucyte images of SytoxGreen-stained stably BV2 cells after doxycycline addition for 24 h. Scale bar, 200 μm. e, BV2 cells expressing inducible constructs of NS3-FL, NS3-N and NS3-C. The time-course Incucyte quantification of SytoxGreen-positive cells in the absence or presence doxycycline. f and g, N-terminal Flag-tagged constructs encoding NS3-FL, NS3-N or NS3-C from MNoVCR6 were transfected into A20 B cells (f) and HeLa cells (g). Cell death was determined by ATP-based cell viability assay 24 h after transfection. h, LDH released from HEK293T cells transfected with N-terminal Flag-tagged constructs encoding NS3-FL, NS3-N or NS3-C of HNoV MD145. i, Representative bright-field images of HEK293T cells transfected with N-terminal Flag-tagged constructs encoding NS3-FL, NS3-N or NS3-C of HNoV MD145. Scale bar, 100 μm. j, The immunoblot shows expression of transfected FL-, N- or C-NS3 of MD145. Data in (a,b,d,e,i and j) are representative of three independent experiments. Data in (c,f,g and h) are pooled from three independent experiments with three technical replicates each. Data are presented as mean of biological replicates ± s.d.

Source Data

Extended Data Fig. 6 Subcellular localization prediction of NS3 and AlphaFold prediction of NS3 and mouse MLKL N-terminal domain.

DeepLoc prediction server revealed that full-length NS3 of (a) HNoV and (b) MNoV have high probability for mitochondrial localization. c, 3D structures of 4HB domain of NS3 (25-158aa) and mouse MLKL (1-125aa) derived from AlphaFold. d, Side views of the structure of mouse MLKL (1-125aa) (yellow), predicted by AlphaFold, was superimposed on mouse MLKL (green) structure previously reported26.

Extended Data Fig. 7 Expanded alignment of cellular and viral 4HB domain sequences.

a, Sequences of cellular MLKL 4HB domains were retrieved from NCBI. Accession numbers are present in Supplemental Table (13). Sequences were aligned using MUSCLE in Geneious Prime. Residues matching consensus are colored. b, NS3 N-terminus amino acid multiple sequence alignment (residues 1-158 relative to MNoV) for thirty-one calicivirus species including representatives from norovirus genogroups. Sequences were retrieved from NCBI. Viral species and strains were selected based on ICTV. Accession numbers are present in Supplemental Table (13). Sequences were aligned using MUSCLE in Geneious Prime. Residues matching consensus are colored.

Extended Data Fig. 8 Expanded alignment of cellular and calicivirus 4HB domain sequences.

Sequences were retrieved from NCBI. Viral species and strains were selected based on ICTV. Accession numbers are present in Supplemental Table. Sequences were aligned using MUSCLE in Geneious Prime. Residues matching consensus are colored.

Extended Data Fig. 9 MNoV infection and expression Full-length or N-terminal NS3 of MNoV altered mitochondria membrane potential and increased mitochondrial ROS abundance.

a, Representative confocal imaging of BV2 cells stably expressing inducible constructs of MNoVCR6 NS3-FL, NS3-N and NS3-C in the presence doxycycline for 10 h. Flag-tagged proteins (green) were detected by immunofluorescence with mitochondrial marker COX IV (red). Scale bar, 10 μm. b, Representative microscopy imaging of NS3 subcellular localization in HEK293T cells. C-terminal Flag-tagged NS3-FL, NS3-N and NS3-C transfected HEK293T cells, the distribution of NS3 (green) was detected by immunofluorescence with cellular membrane marker (yellow). Scale bar, 5 μm. (c–e) Full-length, N- or C-terminal of NS3 fused to a C-terminal Flag tag were stably expressed in BV2 cells under a tetracycline-inducible promoter. Doxycycline was used to induce NS3-FL, NS3-N and NS3-C expression. Cells were first gated off forward and side scatter. c, Flow cytometric analysis of mitochondrial membrane potential (Ψm) in the absence (black) or presence (red) of doxycycline measured by TMRM fluorescence. d, Flow cytometric analysis of mitochondrial status in the absence or presence doxycycline. Gates represent cells with damaged mitochondria. e, Flow cytometric analysis of mitochondrial ROS in the absence (black) or presence (red) of doxycycline. f, Flow cytometric analysis of mitochondrial status in macrophages infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 12 h. Gates represent cells with damaged mitochondria. g, Flow cytometric analysis of mitochondrial ROS in macrophages infected with MNoVCW3 and MNoVCR6 at a MOI of 5 for 12 h. Data in (a,b) are representative of two independent experiments. Data in (c–g) are representative of two independent experiments with three technical replicates of each condition.

Extended Data Fig. 10 Purification of recombinant MNoV NS3-FL and NS3-N, and evidence of self-association and formation of size-selective pores.

a–c, NS3-FL and NS3-N expressing His6-MBP tag were affinity purified by amylose resin. The His6-MBP tag was cleaved and the protein was further purified with Ni2+ resin, followed by size-exclusion chromatography of the purified NS3-FL (a) and NS3-N (b). The second peak of each was collected and Coomassie blue-stained gel (c) is shown. d, Thermal stability shifts measured NS3-FL and NS3-N after purification. e, Purified FL, N- or C-terminal NS3 proteins were incubated with PC liposomes. Liposome leakage was monitored by measuring DPA chelating-induced fluorescence of released Tb3+ relative to that of Triton X-100 treatment. f, Purified NS3-FL protein was incubated with varying sizes of FITC-dextran that was encapsulated in cardiolipin liposomes. Liposome leakage was measured as described in methods. g, Representative agar plates showing transformed E. coli colonies for NS3-FL, NS3-N and NS3-C. h, Bacterial colony-forming units (CFU) per transformation for NS3-FL, NS3-N and NS3-C are shown in the logarithmic form (log10). i, Flag immunoprecipitation of lysates of HEK293T cells, transfected with GFP-NS3-FL, GFP-NS3-N, GFP-NS3-C and/or Flag-NS3-FL, Flag-NS3-N, Flag-NS3-C, were analyzed by immunoblot with indicated antibodies. j, Flag immunoprecipitation of lysates of HEK293T cells, transfected with GFP-NS3-FL and/or Flag-NS3-FL, Flag-NS3-N, Flag-NS3-C, were analyzed by immunoblot with indicated antibodies. k, Oligomerization of NS3 on the liposome membrane. Full length MBP-NS3 proteins were incubated with cardiolipin or OMM liposomes and were subjected to BS3-mediated crosslinking followed by SDS-PAGE gel electrophoresis and Coomassie blue staining. Data in (ae and g) representative of three independent experiments and in (i–k) representative of two independent experiments. Data in (f and h) are pooled from three independent experiments. Data are presented as mean ± s.d.

Source Data

Extended Data Fig. 11 MNoV infection induces externalization of CL.

a, BV2 cells were left untreated, treated with 2 μM staurosporine (STS) for 4 h, or infected with MNoVCW3 and MNoVCR6 (MOI = 1) for 6 h. Mitochondria were isolated and stained with Annexin V-Alexa Fluor 647 and analyzed by flow cytometry. b, Comparison of ATP levels in MNoVCW3 and MNoVCR6 (MOI = 0.1) infected WT and Pls3 siRNA knockdown BMDMs cells for 12 h. c, Pls3 was deleted from BV2 cells by CRISPR genome editing. Lysates of WT or knockout cells were blotted with anti-PLS3 and anti-actin antibodies. d, Representative images of MNoVCW3 and MNoVCR6 (MOI = 0.1) infected WT and Pls3−/− cells for 12 h. Propidium iodide (PI) was added to the cells 20 min before imaging. Scale bar, 100 μm. e, Comparison of ATP levels in MNoVCW3 and MNoVCR6 (MOI = 0.1) infected WT and Pls3−/− cells for 12 h. f, WT and Pls3−/− BV2 cells infected with MNoVCR6 at a MOI = 0.1 for 12 h and supernatant virus was measured by plaque assay. Data in (a,c and d) representative of two independent experiments. Data in (b,e and f) are pooled from three independent experiments with three technical replicates each. Data are presented as mean of biological replicates ± s.d. Data in (f) is analyzed by two-tailed Student’s t-test.

Source Data

Extended Data Fig. 12 Mitochondrial localization of NS3 is essential for inducing cell death and norovirus infection.

a, Representative confocal microscopy images of staining for dsRNA (green) and DAPI (blue) in BMDMs infected with WT (MNoVCW3 and MNoVCR6) or mutant (MNoVCW3ΔN and MNoVCR6ΔN) virus at MOI of 5 for 12 h. Scale bar, 5 μm. b, BMDMs derived from wild-type mice were infected with WT (MNoVCW3 and MNoVCR6) or mutant (MNoVCW3ΔN and MNoVCR6ΔN) viruses. Cells collected at indicated time points and total cell lysates were subjected to anti-NS1 or anti-actin immunoblotting. c, BV2 cells infected with WT (MNoVCR6) and mutant (MNoVCR6ΔN) MNoV at MOI = 1 and viral genomes that were in the supernatant were quantified by qPCR. d, Representative images of Sytox-Green-stained BV2 cells infected with WT (MNoVCR6 and MNoVCW3) and mutant (MNoVCR6ΔN20 and MNoVCW3ΔN20). Scale bar, 100 μm. (e–h) WT mice were challenged with 106 PFU of MNoVCR6 or MNoVCR6ΔN20 perorally. All mice survived infection. Viral genomes were quantified in the (e) MLN, (f) colon, (g) ileum, (h) feces at 7 days post infection. N = 5 for both groups. Dashed line represents the limit of detection. (i) Survival of Stat1−/− mice after challenge with 106 PFU of MNoVCW3 (n = 6) and MNoVCW3ΔN20 (n = 6). Data in (a,b and d) are representative of two independent experiments. Data in (c) pooled from three independent experiments. Data in (eh) pooled from two independent experiments. Data are presented as mean of ± s.d. Statistical analysis was conducted using two-tailed Student’s t-test. P-value for (i) was calculated with a log-rank (Mantel-Cox) test.

Source Data

Supplementary information

Supplementary Information

This file contains Supplementary Fig. 1 with uncropped gel source data and Supplementary Tables 1, 2 and 3.

Reporting Summary

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, G., Zhang, D., Orchard, R.C. et al. Norovirus MLKL-like protein initiates cell death to induce viral egress. Nature 616, 152–158 (2023). https://doi.org/10.1038/s41586-023-05851-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-05851-w

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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