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:

Generation of a live attenuated influenza A vaccine by proteolysis targeting

Abstract

The usefulness of live attenuated virus vaccines has been limited by suboptimal immunogenicity, safety concerns or cumbersome manufacturing processes and techniques. Here we describe the generation of a live attenuated influenza A virus vaccine using proteolysis-targeting chimeric (PROTAC) technology to degrade viral proteins via the endogenous ubiquitin–proteasome system of host cells. We engineered the genome of influenza A viruses in stable cell lines engineered for virus production to introduce a conditionally removable proteasome-targeting domain, generating fully infective PROTAC viruses that were live attenuated by the host protein degradation machinery upon infection. In mouse and ferret models, PROTAC viruses were highly attenuated and able to elicit robust and broad humoral, mucosal and cellular immunity against homologous and heterologous virus challenges. PROTAC-mediated attenuation of viruses may be broadly applicable for generating live attenuated vaccines.

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: Establishment of the PROTAC virus production system.
Fig. 2: Characterization of PROTAC virus M1-PTD.
Fig. 3: Evaluation of the in vivo safety of PROTAC virus M1-PTD in mice and ferrets.
Fig. 4: Evaluation of the immunogenicity of PROTAC virus M1-PTD in mice and ferrets.
Fig. 5: Characterizations of the protective efficacy of M1-PTD in mice and ferrets.

Similar content being viewed by others

Data availability

The gene sequences of WSN influenza virus strain used in this study have been deposited in GenBank under accession numbers CY034138.1, CY034139.1, CY034135.1, CY034134.1, X17336.1, HE802059.1, L25818.1 and CY034136.1. The gene sequences of PR8 influenza virus strain used in this study have been deposited in GenBank under accession numbers CY147541.1, CY147540.1, CY147539.1, CY147534.1, CY147537.1, CY147536.1, CY147535.1 and CY147538.1. 3D structures of M1, PB2, PB1, PA, NP and NS1 have been deposited in the Protein Data Bank (PDB) with PDB IDs: 7JM3, 4WSB, 4WSB, 4IUJ, 2IQH and 4OPH, respectively. Data of VHL expression in human tissues are from GTEx Analysis Release V8 (dbGaP accession number phs000424.v8.p2) (https://gtexportal.org/home/gene/VHL) and also available in the Source Data files. Data of VHL expression in mouse tissues are from RNA sequencing data of E-MTAB-2801 in Expression Atlas and also available in the Source Data files. Data of VHL expression in human lung are from the Human Protein Atlas (http://www.proteinatlas.org/ENSG00000134086-VHL/single+cell+type/lung). All data are available in the manuscript and in Supplementary and Source Data files. No restriction on data availability. Source data are provided with this paper.

References

  1. Yamayoshi, S. & Kawaoka, Y. Current and future influenza vaccines. Nat. Med. 25, 212–220 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Blanco-Lobo, P., Nogales, A., Rodriguez, L. & Martinez-Sobrido, L. Novel approaches for the development of live attenuated influenza vaccines. Viruses 11, 190 (2019).

  3. Coleman, J. R. et al. Virus attenuation by genome-scale changes in codon pair bias. Science 320, 1784–1787 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Mueller, S. et al. Live attenuated influenza virus vaccines by computer-aided rational design. Nat. Biotechnol. 28, 723–726 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Si, L. et al. Generation of influenza A viruses as live but replication-incompetent virus vaccines. Science 354, 1170–1173 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Du, Y. et al. Genome-wide identification of interferon-sensitive mutations enables influenza vaccine design. Science 359, 290–296 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Talon, J. et al. Influenza A and B viruses expressing altered NS1 proteins: a vaccine approach. Proc. Natl Acad. Sci. USA 97, 4309–4314 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mossler, C. et al. Phase I/II trial of a replication-deficient trivalent influenza virus vaccine lacking NS1. Vaccine 31, 6194–6200 (2013).

    Article  PubMed  CAS  Google Scholar 

  9. Steel, J. et al. Live attenuated influenza viruses containing NS1 truncations as vaccine candidates against H5N1 highly pathogenic avian influenza. J. Virol. 83, 1742–1753 (2009).

    Article  CAS  PubMed  Google Scholar 

  10. Wu, C. Y. et al. Influenza A surface glycosylation and vaccine design. Proc. Natl Acad. Sci. USA 114, 280–285 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Wang, L. et al. Generation of a live attenuated influenza vaccine that elicits broad protection in mice and ferrets. Cell Host Microbe 21, 334–343 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Jung, E. J., Lee, K. H. & Seong, B. L. Reverse genetic platform for inactivated and live-attenuated influenza vaccine. Exp. Mol. Med. 42, 116–121 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Krammer, F. & Palese, P. Advances in the development of influenza virus vaccines. Nat. Rev. Drug Discov. 14, 167–182 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Ulmer, J. B., Valley, U. & Rappuoli, R. Vaccine manufacturing: challenges and solutions. Nat. Biotechnol. 24, 1377–1383 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Wei, C. J. et al. Next-generation influenza vaccines: opportunities and challenges. Nat. Rev. Drug Discov. 19, 239–252 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gouma, S., Anderson, E. M. & Hensley, S. E. Challenges of making effective influenza vaccines. Annu. Rev. Virol. 7, 495–512 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Moeller, A., Kirchdoerfer, R. N., Potter, C. S., Carragher, B. & Wilson, I. A. Organization of the influenza virus replication machinery. Science 338, 1631–1634 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Scudellari, M. Protein-slaying drugs could be the next blockbuster therapies. Nature 567, 298–300 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Salami, J. & Crews, C. M. Waste disposal—an attractive strategy for cancer therapy. Science 355, 1163–1167 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Cromm, P. M. & Crews, C. M. Targeted protein degradation: from chemical biology to drug discovery. Cell Chem. Biol. 24, 1181–1190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Deshaies, R. J. Protein degradation: prime time for PROTACs. Nat. Chem. Biol. 11, 634–635 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Yau, R. & Rape, M. The increasing complexity of the ubiquitin code. Nat. Cell Biol. 18, 579–586 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Finley, D. Recognition and processing of ubiquitin–protein conjugates by the proteasome. Annu. Rev. Biochem. 78, 477–513 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hon, W. C. et al. Structural basis for the recognition of hydroxyproline in HIF-1α by pVHL. Nature 417, 975–978 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Jaakkola, P. et al. Targeting of HIF-α to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292, 468–472 (2001).

    Article  CAS  PubMed  Google Scholar 

  26. Ivan, M. et al. HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292, 464–468 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Gu, S., Cui, D., Chen, X., Xiong, X. & Zhao, Y. PROTACs: an emerging targeting technique for protein degradation in drug discovery. Bioessays 40, e1700247 (2018).

    Article  PubMed  CAS  Google Scholar 

  28. Zhang, C. et al. USP9X destabilizes pVHL and promotes cell proliferation. Oncotarget 7, 60519–60534 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Latif, F. et al. Identification of the von Hippel–Lindau disease tumor suppressor gene. Science 260, 1317–1320 (1993).

    Article  CAS  PubMed  Google Scholar 

  30. Iwai, K. et al. Identification of the von Hippel–Lindau tumor-suppressor protein as part of an active E3 ubiquitin ligase complex. Proc. Natl Acad. Sci. USA 96, 12436–12441 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Los, M. et al. Expression pattern of the von Hippel–Lindau protein in human tissues. Lab. Invest. 75, 231–238 (1996).

    CAS  PubMed  Google Scholar 

  32. Yang, J. et al. The I-TASSER Suite: protein structure and function prediction. Nat. Methods 12, 7–8 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Broadbent, A. J., Santos, C. P., Godbout, R. A. & Subbarao, K. The temperature-sensitive and attenuation phenotypes conferred by mutations in the influenza virus PB2, PB1, and NP genes are influenced by the species of origin of the PB2 gene in reassortant viruses derived from influenza A/California/07/2009 and A/WSN/33 viruses. J. Virol. 88, 12339–12347 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Jensen, S. M., Potts, G. K., Ready, D. B. & Patterson, M. J. Specific MHC-I peptides are induced using PROTACs. Front. Immunol. 9, 2697 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Moser, S. C., Voerman, J. S. A., Buckley, D. L., Winter, G. E. & Schliehe, C. Acute pharmacologic degradation of a stable antigen enhances its direct presentation on MHC class I molecules. Front. Immunol. 8, 1920 (2017).

    Article  PubMed  CAS  Google Scholar 

  36. Hussain, A. I., Cordeiro, M., Sevilla, E. & Liu, J. Comparison of egg and high yielding MDCK cell-derived live attenuated influenza virus for commercial production of trivalent influenza vaccine: in vitro cell susceptibility and influenza virus replication kinetics in permissive and semi-permissive cells. Vaccine 28, 3848–3855 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pica, N. & Palese, P. Toward a universal influenza virus vaccine: prospects and challenges. Annu. Rev. Med. 64, 189–202 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Ping, J., Lopes, T. J., Neumann, G. & Kawaoka, Y. Development of high-yield influenza B virus vaccine viruses. Proc. Natl Acad. Sci. USA 113, E8296–E8305 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dolgin, E. mRNA flu shots move into trials. Nat. Rev. Drug Discov. 20, 801–803 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Lowe, D. Moderna’s mRNA flu vaccine. Science https://www.science.org/content/blog-post/moderna-s-mrna-flu-vaccine (2021).

  41. Gossage, L., Eisen, T. & Maher, E. R. VHL, the story of a tumour suppressor gene. Nat. Rev. Cancer 15, 55–64 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Perez Rubio, A. & Eiros, J. M. Cell culture-derived flu vaccine: present and future. Hum. Vaccin. Immunother. 14, 1874–1882 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Takada, A., Matsushita, S., Ninomiya, A., Kawaoka, Y. & Kida, H. Intranasal immunization with formalin-inactivated virus vaccine induces a broad spectrum of heterosubtypic immunity against influenza A virus infection in mice. Vaccine 21, 3212–3218 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Haredy, A. M. et al. An MDCK cell culture-derived formalin-inactivated influenza virus whole-virion vaccine from an influenza virus library confers cross-protective immunity by intranasal administration in mice. Clin. Vaccine Immunol. 20, 998–1007 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Watanabe, S., Watanabe, T. & Kawaoka, Y. Influenza A virus lacking M2 protein as a live attenuated vaccine. J Virol 83, 5947–5950 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Neumann, G. et al. Generation of influenza A viruses entirely from cloned cDNAs. Proc. Natl Acad. Sci. USA 96, 9345–9350 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hoffmann, E., Krauss, S., Perez, D., Webby, R. & Webster, R. G. Eight-plasmid system for rapid generation of influenza virus vaccines. Vaccine 20, 3165–3170 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Anchisi, S., Goncalves, A. R., Mazel-Sanchez, B., Cordey, S. & Schmolke, M. Influenza A virus genetic tools: from clinical sample to molecular clone. Methods Mol. Biol. 1836, 33–58 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Lei, C., Yang, J., Hu, J. & Sun, X. On the calculation of TCID50 for quantitation of virus infectivity. Virol. Sin. 36, 141–144 (2021).

    Article  PubMed  Google Scholar 

  50. Knudson, D. L. & Tinsley, T. W. Replication of a nuclear polyhedrosis virus in a continuous cell culture of Spodoptera frugiperda: purification, assay of infectivity, and growth characteristics of the virus. J. Virol. 14, 934–944 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zheng, W. et al. LOMETS2: improved meta-threading server for fold-recognition and structure-based function annotation for distant-homology proteins. Nucleic Acids Res. 47, W429–W436 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, Y. & Skolnick, J. Scoring function for automated assessment of protein structure template quality. Proteins 57, 702–710 (2004).

  53. Karlsson, M. et al. A single-cell type transcriptomics map of human tissues. Sci. Adv. 7, eabh2169 (2021).

  54. Noda, T. et al. Architecture of ribonucleoprotein complexes in influenza A virus particles. Nature 439, 490–492 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the Animal Experimental Management Center, Public Technology Service Platform, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, for assistance with animal experiments. We acknowledge YuBiolab Ltd., Sinovac Biotech Ltd., TsingKe Biotech Ltd., Genewiz lnc., Corregene Biotech Ltd. and Servicebio Ltd. for technical assistance with animal experiments, gene sequencing, plasmid construction, establishment of cell lines and immunohistochemistry experiments. We thank C. Liu, X. Zhang, S. Huang, Y. Ma, B. Zhang and relevant staffs from the Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, for support of technical and experimental platforms. We thank L. Zhu from the Lung Cancer Center, West China Hospital of Sichuan University, for providing paraffin-embedded and frozen sections of human lung tissue. We acknowledge the Genotype-Tissue Expression (GTEx) Project and/or Portal supported by the Common Fund of the Office of the Director of the National Institutes of Health and by NCI, NHGRI, NHLBI, NIDA, NIMH and NINDS. This work was supported by the National Natural Science Foundation of China (grant to L.S., grant no. 11932014 to X.L., and grant no. 81802456 to H.T.), the China-U.S. Collaborative Program on Emerging and Re-emerging Infectious Diseases (grant no. E2900901-02 to W.T.), Guangdong Basic and Applied Basic Research Foundation (grant no. 2019A1515110615 to H.L.), Natural Science Foundation Program of Shaanxi Province of China (grant no. 2020JQ-545 to H.T.), the Key R&D Program of Shanxi Province of China (grant to W.T.), China Postdoctoral Science Foundation (grant no. 2019TQ0206 to H.L.), and the Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (grant to L.S.).

Author information

Authors and Affiliations

Authors

Contributions

L.S. conceived this study and designed the experiments. L.S., Q.S. and J.L. performed and analyzed experiments, with other authors assisting with experiments and data analysis. L.S. wrote the manuscript, with all authors providing feedback.

Corresponding author

Correspondence to Longlong Si.

Ethics declarations

Competing interests

L.S., Q.S., J.L., L.C., J.S. and X.X. are inventors on the relevant patent applications held by the Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences. The remaining authors declare no competing financial interests.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

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 VHL is widely expressed in tissues of human and mouse.

a, Expression of VHL in human tissues. Data are from GTEx Analysis Release V8 (dbGaP accession number phs000424.v8.p2) (https://gtexportal.org/home/gene/VHL). Box plots are shown as median and 25th and 75th percentiles; points are displayed as outliers if they are above or below 1.5 times the interquartile range. TPM, transcripts per million. b, Expression of VHL in mouse tissues based on RNA-seq data of E-MTAB-2801 in Expression Atlas. Data are presented as mean ± SD; n = 2 for heart and 3 for others. c, Expression of VHL in the human lung. Data are from the Human Protein Atlas (http://www.proteinatlas.org/ENSG00000134086-VHL/single+cell+type/lung)53. pTPM, transcripts per million protein coding genes.

Source data

Extended Data Fig. 2 VHL is widely expressed in lungs of mouse and human.

a, Western blotting images showing the expression of VHL in lungs of BALB/c and C57BL/6 J mice. b, Immunohistochemistry images showing the expression of VHL (brown) in lungs of BALB/c mouse, C57BL/6 J mouse, and human (n = 3). c, Immunofluorescence image showing the expression of VHL in primary human lung alveolar epithelium cultured on Transwells. Green, VHL; blue, nuclei. All experiments were repeated at least two times.

Source data

Extended Data Fig. 3 Schematic structure model of the influenza A virion.

Influenza A virions are enveloped spherical structures with diameters of 80–120 nm54. The genome of each influenza A virion contains eight single-stranded negative-sense RNA segments, encoding 10 main viral proteins: hemagglutinin (HA), neuraminidase (NA), matrix protein 1 (M1), M2, polymerase basic protein 1 (PB1), polymerase basic protein 2 (PB2), polymerase acidic protein (PA), nucleoprotein (NP), nonstructural protein 1 (NS1), and nuclear export protein (NEP, also known as NS2). HA and NA are two main antigenic determinants of influenza virus, which are on the surface of the viral envelope and mediate viral entry into host cells and release of progeny virions from host cells, respectively. M2 is the third integral membrane protein, which forms ion channels in virion particles and has roles in viral entry, assembly, and budding. M1, the most abundant protein in the virion, lines the internal surface of the viral lipid bilayer and mediates the interactions between the viral membrane and ribonucleoprotein (RNP). The RNP is a complex structure composed of multiple copies of the viral NP, which wraps eight viral RNA genome segments, and a single RNA polymerase, which binds to the termini of each RNA genome segment. The RNA polymerase has three subunits: PB1, PB2, and PA, which are responsible for viral replication and transcription17,54. NS1 plays an important role in evasion of the host innate immune system and facilitates viral replication. NEP mediates the export of the newly synthesized viral RNP from the nucleus into the cytoplasm.

Extended Data Fig. 4 Structure prediction of PTD-tagged influenza viral proteins according to I-TASSER32.

Green, viral protein; blue, TEVcs linker; orange, proteasome-targeting peptides. Top images showing the overall structures of influenza viral proteins containing PTD, bottom images showing the close-up views on PTD domain in each influenza viral protein. The PDB IDs of M1, PB2, PB1, PA, NP, and NS1 are 7JM3, 4WSB, 4WSB, 4IUJ, 2IQH, and 4OPH, respectively.

Extended Data Fig. 5 Verification of the genetic stability of M1-PTD by sequencing after 20 passages in MDCK-TEVp cells.

The gene sequence used to construct the M1-PTD was used as the template.

Extended Data Fig. 6 M1-PTD infection had no effect on HIF-1α, the endogenous substrate of VHL, even at MOI = 1.0.

MDCK.2 cells were infected with M1-PTD (MOI = 0, 0.01, 0.1, or 1.0) and analyzed for HIF-1α protein levels by western blotting at 48 h post-infection. The experiment was repeated two times.

Source data

Extended Data Fig. 7 The dose-dependent immune responses elicited by M1-PTD in WT mice and the immune responses elicited by M1-PTD in BALB/c Nude mice.

a, HI antibody responses elicited by indicated dosages of M1-PTD in the sera of WT BALB/c (left) and BALB/c Nude (right) mice at day 21 post-vaccination (n = 5). Data are plotted for individual mice and overlaid with mean ± SD; left graph, one-way ANOVA with Dunnett’s multiple comparisons test; right graph, unpaired two-tailed t-test; n.s., not significant. b, NT antibody responses elicited by indicated dosages of M1-PTD in the sera of WT BALB/c (left) and BALB/c Nude (right) mice at day 21 post-vaccination (n = 5). Data are plotted for individual mice and overlaid with mean ± SD; left graph, one-way ANOVA with Dunnett’s multiple comparisons test; right graph, unpaired two-tailed t-test; n.s., not significant. c, Anti-HA antibody responses elicited by indicated dosages of M1-PTD in the sera of WT BALB/c (left) and BALB/c Nude (right) mice at day 21 post-vaccination (n = 5). Data are plotted for individual mice and overlaid with mean ± SD; left graph, one-way ANOVA with Dunnett’s multiple comparisons test; right graph, unpaired two-tailed t-test; n.s., not significant. d, Anti-NP antibody responses elicited by indicated dosages of M1-PTD in the sera of WT BALB/c (left) and BALB/c Nude (right) mice at day 21 post-vaccination (n = 5). Data are plotted for individual mice and overlaid with mean ± SD; left graph, one-way ANOVA with Dunnett’s multiple comparisons test; right graph, unpaired two-tailed t-test; n.s., not significant. Note that M1-PTD did not induce immune response in T cell-deficient BALB/c Nude mice, indicating that T cell response may be the driver for the immunity elicited by M1-PTD. e, Viral M1 antigen-specific T cell responses elicited by indicated dosages of M1-PTD in the lungs of WT C57BL/6 J mice at day 7 post-vaccination (n = 5), measured by ELISpot assay. Two peptides, M1128-135 MGLIYNRM (left) and M158-66 GILGFVFTL (right), were used as the stimuli. IFNγ-expressing cells per million cells were shown. Data are plotted for individual mice and overlaid with mean ± SD; one-way ANOVA with Dunnett’s multiple comparisons test.

Source data

Extended Data Fig. 8 T cell responses induced by M1-PTD and M1 antigen presentation mediated by PTD.

a, CD8 and CD4 T cell responses elicited by 105 PFU of M1-PTD, CAIV, and M1-KO vaccines in lungs of C57BL/6 J mice at day 7 post-vaccination, measured by flow cytometry. Data are plotted for individual mice and overlaid with mean ± SD; n = 3 mice for Vehicle in left graph, 4 mice for CAIV in left graph, and 5 mice for all other groups; one-way ANOVA with Tukey’s multiple comparisons test. b, Representative flow cytometry histograms are shown for lung samples from mice vaccinated with 105 PFU of M1-PTD. Purple, stained cells; red, unstained cells. c and d, PTD-mediated M1 degradation increases M1 antigen presentation in Raw264.7 cells. Raw264.7 cells were infected with M1-PTD or M1KO virus (MOI = 0.1). 24 hours after infection, M1 antigen presentation on the surface of Raw264.7 cells was detected by an anti-M1 peptide (M1128-135: MGLIYNRM) antibody (c) or an anti-M1 antibody (d) using flow cytometry. Data are presented as mean ± SD; n = 3 biological replicates; unpaired two-tailed t-test.

Source data

Extended Data Fig. 9 Characterization of the broad applicability of PROTAC virus vaccine strategy.

a, Generation and characterization of PROTAC virus M1-PTDPR8 by incorporating PTD to M1 protein of influenza A/Puerto Rico/8/1934 (H1N1) virus (PR8). Characterization of the efficient propagation of M1-PTDPR8 in MDCK-TEVp cells and not in conventional MDCK.2 cells by CPE measured by CellTiter-Glo assay. Data are presented as mean ± SD; n = 3 biological replicates. b, Multicycle replication kinetics curves of M1-PTDPR8 in MDCK-TEVp and conventional MDCK.2 cells. The detection limit is 70 PFU/mL. Data are presented as mean ± SD; n = 3 biological replicates.

Source data

Extended Data Fig. 10 Replication of M1-PTD in TEVp- or viral M1-expressing HEK293T cells.

Relative copy numbers of NP gene of M1-PTD viruses in viral supernatants after 2 days of propagation in TEVp-expressing or viral M1-expressing HEK293T cells indicate that M1-PTD can efficiently replicate in both types of cells. Data are presented as mean ± SD; n = 3 biological replicates.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 1

Unprocessed western blot

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 2

Unprocessed western blots

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Unprocessed western blot

Source Data Extended Data Fig. 6

Unprocessed western blot

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 9

Statistical Source Data

Source Data Extended Data Fig. 10

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Si, L., Shen, Q., Li, J. et al. Generation of a live attenuated influenza A vaccine by proteolysis targeting. Nat Biotechnol 40, 1370–1377 (2022). https://doi.org/10.1038/s41587-022-01381-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41587-022-01381-4

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