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:

Deep sequencing of HIV-1 reverse transcripts reveals the multifaceted antiviral functions of APOBEC3G

Abstract

Following cell entry, the RNA genome of HIV-1 is reverse transcribed into double-stranded DNA that ultimately integrates into the host-cell genome to establish the provirus. These early phases of infection are notably vulnerable to suppression by a collection of cellular antiviral effectors, called restriction or resistance factors. The host antiviral protein APOBEC3G (A3G) antagonizes the early steps of HIV-1 infection through the combined effects of inhibiting viral cDNA production and cytidine-to-uridine-driven hypermutation of this cDNA. In seeking to address the underlying molecular mechanism for inhibited cDNA synthesis, we developed a deep sequencing strategy to characterize nascent reverse transcription products and their precise 3′-termini in HIV-1 infected T cells. Our results demonstrate site- and sequence-independent interference with reverse transcription, which requires the specific interaction of A3G with reverse transcriptase itself. This approach also established, contrary to current ideas, that cellular uracil base excision repair (UBER) enzymes target and cleave A3G-edited uridine-containing viral cDNA. Together, these findings yield further insights into the regulatory interplay between reverse transcriptase, A3G and cellular DNA repair machinery, and identify the suppression of HIV-1 reverse transcriptase by a directly interacting host protein as a new cell-mediated antiviral mechanism.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Effects of A3G on profiles of nascent HIV-1 cDNA products in infected T cells.
Fig. 2: Consequences of UDG inhibition on A3G antiviral phenotype and cDNA profiles.
Fig. 3: Interaction of A3G with HIV-1 RT.
Fig. 4: A3G interaction with HIV-1 RT in virions.
Fig. 5: Mapping of A3G–RT interaction sites on A3G protein.
Fig. 6: Phenotypes of packaged L35A and R24A A3G mutant proteins on viral infectivity and cDNA profiles.

Similar content being viewed by others

References

  1. Doyle, T., Goujon, C. & Malim, M. H. HIV-1 and interferons: who’s interfering with whom? Nat. Rev. Microbiol. 13, 403–413 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Simon, V., Bloch, N. & Landau, N. R. Intrinsic host restrictions to HIV-1 and mechanisms of viral escape. Nat. Immunol. 16, 546–553 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Desimmie, B. A. et al. Multiple APOBEC3 restriction factors for HIV-1 and one Vif to rule them all. J. Mol. Biol. 426, 1220–1245 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Harris, R. S. & Dudley, J. P. APOBECs and virus restriction. Virology 479–480, 131–145 (2015).

    Article  PubMed  Google Scholar 

  5. Bishop, K. N. et al. Cytidine deamination of retroviral DNA by diverse APOBEC proteins. Curr. Biol. 14, 1392–1396 (2004).

    Article  CAS  PubMed  Google Scholar 

  6. 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  PubMed  PubMed Central  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  CAS  PubMed  Google Scholar 

  8. Apolonia, L. et al. Promiscuous RNA binding ensures effective encapsidation of APOBEC3 proteins by HIV-1. PLoS Pathog. 11, e1004609 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Luo, K. et al. Amino-terminal region of the human immunodeficiency virus type 1 nucleocapsid is required for human APOBEC3G packaging. J. Virol. 78, 11841–11852 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Soros, V. B., Yonemoto, W. & Greene, W. C. Newly synthesized APOBEC3G is incorporated into HIV virions, inhibited by HIV RNA, and subsequently activated by RNase H. PLoS Pathog. 3, e15 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Sheehy, A. M., Gaddis, N. C. & Malim, M. H. The antiretroviral enzyme APOBEC3G is degraded by the proteasome in response to HIV-1 Vif. Nat. Med. 9, 1404–1407 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Marin, M., Rose, K. M., Kozak, S. L. & Kabat, D. HIV-1 Vif protein binds the editing enzyme APOBEC3G and induces its degradation. Nat. Med 9, 1398–1403 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Yu, X. et al. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science 302, 1056–1060 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Gillick, K. et al. Suppression of HIV-1 infection by APOBEC3 proteins in primary human CD4+ T cells is associated with inhibition of processive reverse transcription as well as excessive cytidine deamination. J. Virol. 87, 1508–1517 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Phalora, P. K., Sherer, N. M., Wolinsky, S. M., Swanson, C. M. & Malim, M. H. HIV-1 replication and APOBEC3 antiviral activity are not regulated by P bodies. J. Virol. 86, 11712–11724 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Harris, R. S. et al. DNA deamination mediates innate immunity to retroviral infection. Cell 113, 803–809 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Mangeat, B. et al. Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 424, 99–103 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Zhang, H. et al. The cytidine deaminase CEM15 induces hypermutation in newly synthesized HIV-1 DNA. Nature 424, 94–98 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bishop, K. N., Holmes, R. K. & Malim, M. H. Antiviral potency of APOBEC proteins does not correlate with cytidine deamination. J. Virol. 80, 8450–8458 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bishop, K. N., Verma, M., Kim, E. Y., Wolinsky, S. M. & Malim, M. H. APOBEC3G inhibits elongation of HIV-1 reverse transcripts. PLoS Pathog. 4, e1000231 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Holmes, R. K., Koning, F. A., Bishop, K. N. & Malim, M. H. APOBEC3F can inhibit the accumulation of HIV-1 reverse transcription products in the absence of hypermutation. Comparisons with APOBEC3G. J. Biol. Chem. 282, 2587–2595 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Iwatani, Y. et al. Deaminase-independent inhibition of HIV-1 reverse transcription by APOBEC3G. Nucleic Acids Res. 35, 7096–7108 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Schrofelbauer, B., Yu, Q., Zeitlin, S. G. & Landau, N. R. Human immunodeficiency virus type 1 Vpr induces the degradation of the UNG and SMUG uracil-DNA glycosylases. J. Virol. 79, 10978–10987 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, B., Chen, K., Zhang, C., Huang, S. & Zhang, H. Virion-associated uracil DNA glycosylase-2 and apurinic/apyrimidinic endonuclease are involved in the degradation of APOBEC3G-edited nascent HIV-1 DNA. J. Biol. Chem. 282, 11667–11675 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Kaiser, S. M. & Emerman, M. Uracil DNA glycosylase is dispensable for human immunodeficiency virus type 1 replication and does not contribute to the antiviral effects of the cytidine deaminase Apobec3G. J. Virol. 80, 875–882 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Langlois, M. A. & Neuberger, M. S. Human APOBEC3G can restrict retroviral infection in avian cells and acts independently of both UNG and SMUG1. J. Virol. 82, 4660–4664 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mbisa, J. L. et al. Human immunodeficiency virus type 1 cDNAs produced in the presence of APOBEC3G exhibit defects in plus-strand DNA transfer and integration. J. Virol. 81, 7099–7110 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Newman, E. N. et al. Antiviral function of APOBEC3G can be dissociated from cytidine deaminase activity. Curr. Biol. 15, 166–170 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Mbisa, J. L., Bu, W. & Pathak, V. K. APOBEC3F and APOBEC3G inhibit HIV-1 DNA integration by different mechanisms. J. Virol. 84, 5250–5259 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chaurasiya, K. R. et al. Oligomerization transforms human APOBEC3G from an efficient enzyme to a slowly dissociating nucleic acid-binding protein. Nat. Chem. 6, 28–33 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Wang, X. et al. The cellular antiviral protein APOBEC3G interacts with HIV-1 reverse transcriptase and inhibits its function during viral replication. J. Virol. 86, 3777–3786 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hu, W. S. & Hughes, S. H. HIV-1 reverse transcription. C.S.H. Perspect. Med. 2, a006882 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Herschhorn, A. & Hizi, A. Retroviral reverse transcriptases. Cell Mol. Life Sci. 67, 2717–2747 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Arts, E. J., Li, Z. & Wainberg, M. A. Analysis of primer extension and the first template switch during human immunodeficiency virus reverse transcription. J. Biomed. Sci. 2, 314–321 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Driscoll, M. D., Golinelli, M. P. & Hughes, S. H. In vitro analysis of human immunodeficiency virus type 1 minus-strand strong-stop DNA synthesis and genomic RNA processing. J. Virol. 75, 672–686 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Harrison, G. P., Mayo, M. S., Hunter, E. & Lever, A. M. Pausing of reverse transcriptase on retroviral RNA templates is influenced by secondary structures both 5′ and 3′ of the catalytic site. Nucleic Acids Res. 26, 3433–3442 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kim, E. Y. et al. Human APOBEC3 induced mutation of human immunodeficiency virus type-1 contributes to adaptation and evolution in natural infection. PLoS Pathog. 10, e1004281 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Yu, Q. et al. Single-strand specificity of APOBEC3G accounts for minus-strand deamination of the HIV genome. Nat. Struct. Mol. Biol. 11, 435–442 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Schormann, N., Ricciardi, R. & Chattopadhyay, D. Uracil-DNA glycosylases—structural and functional perspectives on an essential family of DNA repair enzymes. Protein Sci. 23, 1667–1685 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Visnes, T. et al. Uracil in DNA and its processing by different DNA glycosylases. Philos. T. Roy. Soc. B. 364, 563–568 (2009).

    Article  CAS  Google Scholar 

  41. Wang, Z. & Mosbaugh, D. W. Uracil-DNA glycosylase inhibitor gene of bacteriophage PBS2 encodes a binding protein specific for uracil-DNA glycosylase. J. Biol. Chem. 264, 1163–1171 (1989).

    CAS  PubMed  Google Scholar 

  42. Mansky, L. M., Preveral, S., Selig, L., Benarous, R. & Benichou, S. The interaction of vpr with uracil DNA glycosylase modulates the human immunodeficiency virus type 1 in vivo mutation rate. J. Virol. 74, 7039–7047 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Willetts, K. E. et al. DNA repair enzyme uracil DNA glycosylase is specifically incorporated into human immunodeficiency virus type 1 viral particles through a Vpr-independent mechanism. J. Virol. 73, 1682–1688 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Adolph, M. B., Webb, J. & Chelico, L. Retroviral restriction factor APOBEC3G delays the initiation of DNA synthesis by HIV-1 reverse transcriptase. PLoS ONE 8, e64196 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Voss, T. C., Demarco, I. A. & Day, R. N. Quantitative imaging of protein interactions in the cell nucleus. Biotechniques 38, 413–424 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Burnett, A. & Spearman, P. APOBEC3G multimers are recruited to the plasma membrane for packaging into human immunodeficiency virus type 1 virus-like particles in an RNA-dependent process requiring the NC basic linker. J. Virol. 81, 5000–5013 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wiegand, H. L., Doehle, B. P., Bogerd, H. P. & Cullen, B. R. A second human antiretroviral factor, APOBEC3F, is suppressed by the HIV-1 and HIV-2 Vif proteins. EMBO J. 23, 2451–2458 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wedekind, J. E. et al. Nanostructures of APOBEC3G support a hierarchical assembly model of high molecular mass ribonucleoprotein particles from dimeric subunits. J. Biol. Chem. 281, 38122–38126 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gallois-Montbrun, S. et al. Comparison of cellular ribonucleoprotein complexes associated with the APOBEC3F and APOBEC3G antiviral proteins. J. Virol. 82, 5636–5642 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Huthoff, H., Autore, F., Gallois-Montbrun, S., Fraternali, F. & Malim, M. H. RNA-dependent oligomerization of APOBEC3G is required for restriction of HIV-1. PLoS Pathog. 5, e1000330 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Xiao, X., Li, S. X., Yang, H. & Chen, X. S. Crystal structures of APOBEC3G N-domain alone and its complex with DNA. Nat. Commun. 7, 12193 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Menendez-Arias, L., Sebastian-Martin, A. & Alvarez, M. Viral reverse transcriptases. Virus Res. 15, 153–176 (2017).

    Article  Google Scholar 

  53. Levin, J. G., Mitra, M., Mascarenhas, A. & Musier-Forsyth, K. Role of HIV-1 nucleocapsid protein in HIV-1 reverse transcription. RNA Biol. 7, 754–774 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Basu, V. P. et al. Strand transfer events during HIV-1 reverse transcription. Virus Res. 134, 19–38 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Iwatani, Y., Rosen, A. E., Guo, J., Musier-Forsyth, K. & Levin, J. G. Efficient initiation of HIV-1 reverse transcription in vitro. Requirement for RNA sequences downstream of the primer binding site abrogated by nucleocapsid protein-dependent primer–template interactions. J. Biol. Chem. 278, 14185–14195 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Masuda, T. et al. Fate of HIV-1 cDNA intermediates during reverse transcription is dictated by transcription initiation site of virus genomic RNA. Sci. Rep. 5, 17680 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kharytonchyk, S. et al. Transcriptional start site heterogeneity modulates the structure and function of the HIV-1 genome. Proc. Natl Acad. Sci. USA 113, 13378–13383 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Klarmann, G. J., Schauber, C. A. & Preston, B. D. Template-directed pausing of DNA synthesis by HIV-1 reverse transcriptase during polymerization of HIV-1 sequences in vitro. J. Biol. Chem. 268, 9793–9802 (1993).

    CAS  PubMed  Google Scholar 

  59. Altfeld, M. & Gale, M. Jr. Innate immunity against HIV-1 infection. Nat. Immunol. 16, 554–562 (2015).

    Article  CAS  PubMed  Google Scholar 

  60. Ahn, J. et al. HIV-1 Vpr loads uracil DNA glycosylase-2 onto DCAF1, a substrate recognition subunit of a cullin 4A-ring E3 ubiquitin ligase for proteasome-dependent degradation. J. Biol. Chem. 285, 37333–37341 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Steagall, W. K., Robek, M. D., Perry, S. T., Fuller, F. J. & Payne, S. L. Incorporation of uracil into viral DNA correlates with reduced replication of EIAV in macrophages. Virology 210, 302–313 (1995).

    Article  CAS  PubMed  Google Scholar 

  62. Kennedy, E. M. et al. Abundant non-canonical dUTP found in primary human macrophages drives its frequent incorporation by HIV-1 reverse transcriptase. J. Biol. Chem. 286, 25047–25055 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Yan, N., O’Day, E., Wheeler, L. A., Engelman, A. & Lieberman, J. HIV DNA is heavily uracilated, which protects it from autointegration. Proc. Natl Acad. Sci. USA 108, 9244–9249 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hansen, E. C. et al. Diverse fates of uracilated HIV-1 DNA during infection of myeloid lineage cells. eLife 5, 18447 (2016).

    Article  Google Scholar 

  65. Weil, A. F. et al. Uracil DNA glycosylase initiates degradation of HIV-1 cDNA containing misincorporated dUTP and prevents viral integration. Proc. Natl Acad. Sci. USA 110, E448–E457 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Chen, R., Wang, H. & Mansky, L. M. Roles of uracil-DNA glycosylase and dUTPase in virus replication. J. Gen. Virol. 83, 2339–2345 (2002).

    Article  CAS  PubMed  Google Scholar 

  67. Sire, J., Querat, G., Esnault, C. & Priet, S. Uracil within DNA: an actor of antiviral immunity. Retrovirology 5, 45 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Stremlau, M. et al. The cytoplasmic body component TRIM5α restricts HIV-1 infection in Old World monkeys. Nature 427, 848–853 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Stremlau, M. et al. Specific recognition and accelerated uncoating of retroviral capsids by the TRIM5α restriction factor. Proc. Natl Acad. Sci. USA 103, 5514–5519 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Malim, M. H. & Bieniasz, P. D. HIV restriction factors and mechanisms of evasion. C.S.H. Perspect. Med. 2, a006940 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Warren, K., Warrilow, D., Meredith, L. & Harrich, D. Reverse transcriptase and cellular factors: regulators of HIV-1 reverse transcription. Viruses 1, 873–894 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Salter, J. D., Bennett, R. P. & Smith, H. C. The APOBEC protein family: united by structure, divergent in function. Trends Biochem Sci 41, 578–594 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Huthoff, H. & Malim, M. H. Identification of amino acid residues in APOBEC3G required for regulation by human immunodeficiency virus type 1 Vif and virion encapsidation. J. Virol. 81, 3807–3815 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Swanson, C. M., Sherer, N. M. & Malim, M. H. SRp40 and SRp55 promote the translation of unspliced human immunodeficiency virus type 1 RNA. J. Virol. 84, 6748–6759 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Gallois-Montbrun, S. et al. Antiviral protein APOBEC3G localizes to ribonucleoprotein complexes found in P bodies and stress granules. J. Virol. 81, 2165–2178 (2007).

    Article  CAS  PubMed  Google Scholar 

  76. Gaddis, N. C., Chertova, E., Sheehy, A. M., Henderson, L. E. & Malim, M. H. Comprehensive investigation of the molecular defect in vif-deficient human immunodeficiency virus type 1 virions. J. Virol. 77, 5810–5820 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Myszka, D. G. Improving biosensor analysis. J. Mol. Recognit. 12, 279–284 (1999).

    Article  CAS  PubMed  Google Scholar 

  78. Chen, Y., Mills, J. D. & Periasamy, A. Protein localization in living cells and tissues using FRET and FLIM. Differentiation 71, 528–541 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Wallrabe, H. & Periasamy, A. Imaging protein molecules using FRET and FLIM microscopy. Curr. Opin. Biotechnol. 16, 19–27 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Becker, W. Fluorescence lifetime imaging—techniques and applications. J. Microsc. 247, 119–136 (2012).

    Article  CAS  PubMed  Google Scholar 

  81. Boyer, P. L., Clark, P. K. & Hughes, S. H. HIV-1 and HIV-2 reverse transcriptases: different mechanisms of resistance to nucleoside reverse transcriptase inhibitors. J. Virol. 86, 5885–5894 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Boyer, P. L., Ding, J., Arnold, E. & Hughes, S. H. Subunit specificity of mutations that confer resistance to nonnucleoside inhibitors in human immunodeficiency virus type 1 reverse transcriptase. Antimicrob. Agents Chemother. 38, 1909–1914 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Peter, M. et al. Multiphoton-FLIM quantification of the EGFP-mRFP1 FRET pair for localization of membrane receptor-kinase interactions. Biophys. J. 88, 1224–1237 (2005).

    Article  CAS  PubMed  Google Scholar 

  84. Parsons, M. et al. Spatially distinct binding of Cdc42 to PAK1 and N-WASP in breast carcinoma cells. Mol. Cell. Biol. 25, 1680–1695 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Prag, S. et al. Activated ezrin promotes cell migration through recruitment of the GEF Dbl to lipid rafts and preferential downstream activation of Cdc42. Mol. Biol. Cell 18, 2935–2948 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Iwatani, Y., Takeuchi, H., Strebel, K. & Levin, J. G. Biochemical activities of highly purified, catalytically active human APOBEC3G: correlation with antiviral effect. J. Virol. 80, 5992–6002 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Bruns, A. M. et al. ATP hydrolysis enhances RNA recognition and antiviral signal transduction by the innate immune sensor, laboratory of genetics and physiology 2 (LGP2). J. Biol. Chem. 288, 938–946 (2013).

    Article  CAS  PubMed  Google Scholar 

  88. Hwang, H. & Myong, S. Protein induced fluorescence enhancement (PIFE) for probing protein–nucleic acid interactions. Chem. Soc. Rev. 43, 1221–1229 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Myong, S. et al. Cytosolic viral sensor RIG-I is a 5′-triphosphate-dependent translocase on double-stranded RNA. Science 323, 1070–1074 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Petersen-Mahrt, S. K., Harris, R. S. & Neuberger, M. S. AID mutates E. coli suggesting a DNA deamination mechanism for antibody diversification. Nature 418, 99–103 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge support (and debate) from members of the Malim laboratory, the insights of J. Ule and R. Oakey on optimization of the sequencing protocol, and M. Emerman and S. Hughes for the provision of reagents. The authors thank M. Arno at the King’s College London Genomic Centre and D. Hughes at the University College London (UCL) Institute for Neurology Next Generation Sequencing Facility for help with MiSeq sequencing runs. The work was supported by the UK Medical Research Council (G1000196 and MR/M001199/1 to M.M. and MR/K015664/1 to M.P.), the Wellcome Trust (106223/Z/14/Z to M.M.), the European Commission’s Seventh Framework Programme (FP7/2007-2013) under grant agreement no. PIIF-GA-2012-329679 (to D.P.), King’s alumni community sponsored King’s Undergraduate Research Fellowships (to R.D.L.), King’s School of Medicine Summer Studentship Award (to J.C.) and the Department of Health via a National Institutes for Health Research Comprehensive Biomedical Research Center award to Guy’s and St. Thomas’ NHS Foundation Trust in partnership with King’s College London and King’s College Hospital NHS Foundation Trust (guysbrc-2012-1).

Author information

Authors and Affiliations

Authors

Contributions

D.P. co-wrote the manuscript and executed all experiments with the following exceptions. R.D.L. performed the co-immunoprecipitation shown in Fig. 5a, and M.P. carried out all the microscopy and FRET-FLIM experiments. A.E.S. wrote and ran the analysis software for analysing raw FASTQ sequencing data. S.C. carried out the double alanine scan for the A3G–RT binding site mapping. A.M.B. and C.M.H. carried out and analysed the single-molecule RNA binding assays (Supplementary Fig. 3c). S.P., R.D.L. and J.A.C. contributed to reagent generation, in particular for Fig. 5. J.M.M. contributed to the SPR experiments (Fig. 3c) and performed the analysis. L.A. and A.E.S. contributed to the sequencing library design. D.P. and M.H.M. conceived the experiments and co-wrote the manuscript. All authors cross-checked the manuscript.

Corresponding author

Correspondence to Michael H. Malim.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

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

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pollpeter, D., Parsons, M., Sobala, A.E. et al. Deep sequencing of HIV-1 reverse transcripts reveals the multifaceted antiviral functions of APOBEC3G. Nat Microbiol 3, 220–233 (2018). https://doi.org/10.1038/s41564-017-0063-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-017-0063-9

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