Vif hijacks CBF-β to degrade APOBEC3G and promote HIV-1 infection (original) (raw)

Nature volume 481, pages 371–375 (2012)Cite this article

Subjects

Abstract

Restriction factors, such as the retroviral complementary DNA deaminase APOBEC3G, are cellular proteins that dominantly block virus replication1,2,3. The AIDS virus, human immunodeficiency virus type 1 (HIV-1), produces the accessory factor Vif, which counteracts the host’s antiviral defence by hijacking a ubiquitin ligase complex, containing CUL5, ELOC, ELOB and a RING-box protein, and targeting APOBEC3G for degradation4,5,6,7,8,9,10. Here we reveal, using an affinity tag/purification mass spectrometry approach, that Vif additionally recruits the transcription cofactor CBF-β to this ubiquitin ligase complex. CBF-β, which normally functions in concert with RUNX DNA binding proteins, allows the reconstitution of a recombinant six-protein assembly that elicits specific polyubiquitination activity with APOBEC3G, but not the related deaminase APOBEC3A. Using RNA knockdown and genetic complementation studies, we also demonstrate that CBF-β is required for Vif-mediated degradation of APOBEC3G and therefore for preserving HIV-1 infectivity. Finally, simian immunodeficiency virus (SIV) Vif also binds to and requires CBF-β to degrade rhesus macaque APOBEC3G, indicating functional conservation. Methods of disrupting the CBF-β–Vif interaction might enable HIV-1 restriction and provide a supplement to current antiviral therapies that primarily target viral proteins.

This is a preview of subscription content, access via your institution

Access options

Subscribe to this journal

Receive 51 print issues and online access

$199.00 per year

only $3.90 per issue

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Additional access options:

Similar content being viewed by others

References

  1. Goff, S. P. Retrovirus restriction factors. Mol. Cell 16, 849–859 (2004)
    Article CAS Google Scholar
  2. Malim, M. H. & Emerman, M. HIV-1 accessory proteins—ensuring viral survival in a hostile environment. Cell Host Microbe 3, 388–398 (2008)
    Article CAS Google Scholar
  3. Albin, J. S. & Harris, R. S. Interactions of host APOBEC3 restriction factors with HIV-1 in vivo: implications for therapeutics. Expert Rev. Mol. Med. 12, e4 (2010)
    Article Google Scholar
  4. Yu, X. et al. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science 302, 1056–1060 (2003)
    Article ADS CAS Google Scholar
  5. Mehle, A., Goncalves, J., Santa-Marta, M., McPike, M. & Gabuzda, D. Phosphorylation of a novel SOCS-box regulates assembly of the HIV-1 Vif-Cul5 complex that promotes APOBEC3G degradation. Genes Dev. 18, 2861–2866 (2004)
    Article CAS Google Scholar
  6. Mehle, A. et al. Vif overcomes the innate antiviral activity of APOBEC3G by promoting its degradation in the ubiquitin-proteasome pathway. J. Biol. Chem. 279, 7792–7798 (2004)
    Article CAS Google Scholar
  7. Sheehy, A. M., Gaddis, N. C., Choi, J. D. & Malim, M. H. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature 418, 646–650 (2002)
    Article ADS CAS Google Scholar
  8. Sheehy, A. M., Gaddis, N. C. & Malim, M. H. The antiretroviral enzyme APOBEC3G is degraded by the proteasome in response to HIV-1 Vif. Nature Med. 9, 1404–1407 (2003)
    Article CAS Google Scholar
  9. Conticello, S. G., Harris, R. S. & Neuberger, M. S. The Vif protein of HIV triggers degradation of the human antiretroviral DNA deaminase APOBEC3G. Curr. Biol. 13, 2009–2013 (2003)
    Article CAS Google Scholar
  10. Stopak, K., de Noronha, C., Yonemoto, W. & Greene, W. C. HIV-1 Vif blocks the antiviral activity of APOBEC3G by impairing both its translation and intracellular stability. Mol. Cell 12, 591–601 (2003)
    Article CAS Google Scholar
  11. Passmore, L. A. & Barford, D. Getting into position: the catalytic mechanisms of protein ubiquitylation. Biochem. J. 379, 513–525 (2004)
    Article CAS Google Scholar
  12. Thrower, J. S., Hoffman, L., Rechsteiner, M. & Pickart, C. M. Recognition of the polyubiquitin proteolytic signal. EMBO J. 19, 94–102 (2000)
    Article CAS Google Scholar
  13. Stanley, B. J. et al. Structural insight into the human immunodeficiency virus Vif SOCS box and its role in human E3 ubiquitin ligase assembly. J. Virol. 82, 8656–8663 (2008)
    Article CAS Google Scholar
  14. Jäger, S. et al. Purification and characterization of HIV-human protein complexes. Methods 53, 13–19 (2011)
    Article Google Scholar
  15. Jäger, S. et al. Global landscape of HIV–human protein complexes. Nature doi:10.1038/nature10719 (this issue).
  16. Kamura, T. et al. VHL-box and SOCS-box domains determine binding specificity for Cul2-Rbx1 and Cul5-Rbx2 modules of ubiquitin ligases. Genes Dev. 18, 3055–3065 (2004)
    Article CAS Google Scholar
  17. Huang, D. T. et al. E2-RING expansion of the NEDD8 cascade confers specificity to cullin modification. Mol. Cell 33, 483–495 (2009)
    Article CAS Google Scholar
  18. Wong, W. F., Kohu, K., Chiba, T., Sato, T. & Satake, M. Interplay of transcription factors in T-cell differentiation and function: the role of Runx. Immunology 132, 157–164 (2011)
    Article CAS Google Scholar
  19. Petroski, M. D. & Deshaies, R. J. Mechanism of lysine 48-linked ubiquitin-chain synthesis by the cullin-RING ubiquitin-ligase complex SCF-Cdc34. Cell 123, 1107–1120 (2005)
    Article CAS Google Scholar
  20. Kim, H. T. et al. Certain pairs of ubiquitin-conjugating enzymes (E2s) and ubiquitin-protein ligases (E3s) synthesize nondegradable forked ubiquitin chains containing all possible isopeptide linkages. J. Biol. Chem. 282, 17375–17386 (2007)
    Article CAS Google Scholar
  21. DeHart, J. L., Bosque, A., Harris, R. S. & Planelles, V. Human immunodeficiency virus type 1 Vif induces cell cycle delay via recruitment of the same E3 ubiquitin ligase complex that targets APOBEC3 proteins for degradation. J. Virol. 82, 9265–9272 (2008)
    Article CAS Google Scholar
  22. Stenglein, M. D., Burns, M. B., Li, M., Lengyel, J. & Harris, R. S. APOBEC3 proteins mediate the clearance of foreign DNA from human cells. Nature Struct. Mol. Biol. 17, 222–229 (2010)
    Article CAS Google Scholar
  23. Hultquist, J. F. et al. Human and rhesus APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H demonstrate a conserved capacity to restrict Vif-deficient HIV-1. J. Virol. 85, 11220–11234 (2011)
    Article CAS Google Scholar
  24. Rodrigo-Brenni, M. C. & Morgan, D. O. Sequential E2s drive polyubiquitin chain assembly on APC targets. Cell 130, 127–139 (2007)
    Article CAS Google Scholar
  25. Wu, K., Kovacev, J. & Pan, Z. Q. Priming and extending: a UbcH5/Cdc34 E2 handoff mechanism for polyubiquitination on a SCF substrate. Mol. Cell 37, 784–796 (2010)
    Article CAS Google Scholar
  26. Koning, F. A. et al. Defining APOBEC3 expression patterns in human tissues and hematopoietic cell subsets. J. Virol. 83, 9474–9485 (2009)
    Article CAS Google Scholar
  27. Refsland, E. W. et al. Quantitative profiling of the full APOBEC3 mRNA repertoire in lymphocytes and tissues: implications for HIV-1 restriction. Nucleic Acids Res. 38, 4274–4284 (2010)
    Article CAS Google Scholar
  28. Saha, A. & Deshaies, R. J. Multimodal activation of the ubiquitin ligase SCF by Nedd8 conjugation. Mol. Cell 32, 21–31 (2008)
    Article CAS Google Scholar

Download references

Acknowledgements

We thank members of the Krogan, Gross and Harris laboratories for comments, J. R. Johnson for mass spectrometry, B. Leonard for sharing unpublished data, M. Shales for help with figures, and B. Chesebro, D. Gabuzda, J. Lingappa, M. Malim, K. Wehrly, X. F. Yu, A. Bullock, B. Schulman and the AIDS Research and Reference Reagent Program for reagents. This research was funded by grants from QB3 at University of California, San Francisco, and the National Institutes of Health (P50 GM082250, P01 AI090935 and P50 GM081879 to N.J.K.; U54 RR022220 to A.S.; R01 AI064046 and P01 GM091743 to R.S.H.; P50 GM082250 to J.D.G. and C.S.C.; P41RR001614 and P50GM081879 to A.B.). N.J.K. is a Searle Scholar and a Keck Young Investigator.

Author information

Author notes

  1. Stefanie Jäger, Dong Young Kim and Judd F. Hultquist: These authors contributed equally to this work.

Authors and Affiliations

  1. Department of Cellular and Molecular Pharmacology, University of California-San Francisco, San Francisco, 94158, California, USA
    Stefanie Jäger, Cathal Mahon, Joshua Kane, Kathy Franks-Skiba & Nevan J. Krogan
  2. California Institute for Quantitative Biosciences, QB3, San Francisco, California, California 94158, USA ,
    Stefanie Jäger, Cathal Mahon, Joshua Kane, Kathy Franks-Skiba, Peter Cimermancic, Alma Burlingame, Andrej Sali, Charles S. Craik, John D. Gross & Nevan J. Krogan
  3. Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158, California, USA
    Dong Young Kim, Eunju Kwon, Linda Yen, David Stanley, Cathal Mahon, Alma Burlingame, Andrej Sali, Charles S. Craik & John D. Gross
  4. Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, University of Minnesota, Minneapolis, 55455, Minnesota, USA
    Judd F. Hultquist, Keisuke Shindo, Rebecca S. LaRue, Ming Li, Brett D. Anderson & Reuben S. Harris
  5. Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 94158, California, USA
    Peter Cimermancic & Andrej Sali
  6. HPC (Host Pathogen Circuitry) Group, University of California-San Francisco, San Francisco, 94158, California, USA
    Andrej Sali, John D. Gross & Nevan J. Krogan
  7. J. David Gladstone Institutes, San Francisco, 94158, California, USA
    Nevan J. Krogan

Authors

  1. Stefanie Jäger
    You can also search for this author inPubMed Google Scholar
  2. Dong Young Kim
    You can also search for this author inPubMed Google Scholar
  3. Judd F. Hultquist
    You can also search for this author inPubMed Google Scholar
  4. Keisuke Shindo
    You can also search for this author inPubMed Google Scholar
  5. Rebecca S. LaRue
    You can also search for this author inPubMed Google Scholar
  6. Eunju Kwon
    You can also search for this author inPubMed Google Scholar
  7. Ming Li
    You can also search for this author inPubMed Google Scholar
  8. Brett D. Anderson
    You can also search for this author inPubMed Google Scholar
  9. Linda Yen
    You can also search for this author inPubMed Google Scholar
  10. David Stanley
    You can also search for this author inPubMed Google Scholar
  11. Cathal Mahon
    You can also search for this author inPubMed Google Scholar
  12. Joshua Kane
    You can also search for this author inPubMed Google Scholar
  13. Kathy Franks-Skiba
    You can also search for this author inPubMed Google Scholar
  14. Peter Cimermancic
    You can also search for this author inPubMed Google Scholar
  15. Alma Burlingame
    You can also search for this author inPubMed Google Scholar
  16. Andrej Sali
    You can also search for this author inPubMed Google Scholar
  17. Charles S. Craik
    You can also search for this author inPubMed Google Scholar
  18. Reuben S. Harris
    You can also search for this author inPubMed Google Scholar
  19. John D. Gross
    You can also search for this author inPubMed Google Scholar
  20. Nevan J. Krogan
    You can also search for this author inPubMed Google Scholar

Contributions

K.S., R.S.L. and E.K. made equal secondary contributions to this work. S.J. and K.F.S. generated the Vif protein–protein interaction map (Fig. 1); S.J. performed the co-immunoprecipitation confirmation assays (Supplementary Fig. 1); P.C. developed and implemented the MiST scoring system (Fig. 1c); S.J. and E.K. performed double purification analyses (Fig. 1c, d); C.M. and J.K. performed immunoprecipitation analyses (Fig. 1f, g); D.Y.K., L.Y. and D.S. reconstituted the Vif E3 ligase from recombinant components and performed ubiquitination assays (Fig. 2 and Supplementary Figs 2–4); M.L. expressed and purified A3 proteins and did in vitro pulldowns (Fig. 2); K.S., J.F.H. and R.S.L. performed CBF-β knockdown, complementation and virus infectivity experiments (Fig. 3 and Supplementary Figs 5–7); and B.D.A. and J.F.H. did the CBF-β–Vif co-immunoprecipitation experiments (Supplementary Fig. 8). A.B., A.S., C.S.C., R.S.H., J.D.G. and N.J.K. supervised the research; S.J., D.Y.K., J.F.H., K.S., R.S.H., J.D.G. and N.J.K. wrote and revised the manuscript.

Corresponding authors

Correspondence toReuben S. Harris, John D. Gross or Nevan J. Krogan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

The file contains Supplementary Methods, Supplementary References and Supplementary Figures 1-8 with legends. (PDF 1666 kb)

PowerPoint slides

Rights and permissions

About this article

Cite this article

Jäger, S., Kim, D., Hultquist, J. et al. Vif hijacks CBF-β to degrade APOBEC3G and promote HIV-1 infection.Nature 481, 371–375 (2012). https://doi.org/10.1038/nature10693

Download citation

This article is cited by

Editorial Summary

Vif–CBF-β interaction an anti-HIV-1 target

The transcription cofactor CBF-β (core binding factor β) regulates the DNA binding activity of RUNX family proteins. Two independent studies now show that CBF-β also regulates the ability of HIV-1 to evade host restriction mediated by the cDNA deaminase APOBEC3G, a host factor that blocks viral replication. They show that it associates with the HIV protein Vif, and is essential for the assembly of the Vif-Cul5 E3 ubiquitin ligase complex, which mediates the ubiquitination and destruction of APOBEC3. Both groups suggest that disrupting the Vif–CBF-β interaction could provide a new therapeutic target against HIV-1 infection.