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:

Vpu modulates DNA repair to suppress innate sensing and hyper-integration of HIV-1

Abstract

To avoid innate sensing and immune control, human immunodeficiency virus type 1 (HIV-1) has to prevent the accumulation of viral complementary DNA species. Here, we show that the late HIV-1 accessory protein Vpu hijacks DNA repair mechanisms to promote degradation of nuclear viral cDNA in cells that are already productively infected. Vpu achieves this by interacting with RanBP2–RanGAP1*SUMO1–Ubc9 SUMO E3-ligase complexes at the nuclear pore to reprogramme promyelocytic leukaemia protein nuclear bodies and reduce SUMOylation of Bloom syndrome protein, unleashing end degradation of viral cDNA. Concomitantly, Vpu inhibits RAD52-mediated homologous repair of viral cDNA, preventing the generation of dead-end circular forms of single copies of the long terminal repeat and permitting sustained nucleolytic attack. Our results identify Vpu as a key modulator of the DNA repair machinery. We show that Bloom syndrome protein eliminates nuclear HIV-1 cDNA and thereby suppresses immune sensing and proviral hyper-integration. Therapeutic targeting of DNA repair may facilitate the induction of antiviral immunity and suppress proviral integration replenishing latent HIV reservoirs.

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: Proviral HIV-1 gene expression modulates homologous repair.
Fig. 2: Viral proteins involved in HIV-1-modulated homologous repair.
Fig. 3: HIV-1 regulates BLM-mediated DNA-end processing.
Fig. 4: Effects of Vpu on SUMOylation and PML-NBs.
Fig. 5: Interaction of Vpu with RanBP2–RanGAP1*SUMO1–Ubc9 SUMO E3-ligase complexes.
Fig. 6: Modulation of viral DNA species and superinfection by Vpu, RAD52 and BLM.

Similar content being viewed by others

Data availability

The data that support the finding of this study are available from the corresponding author on request. Complete western blot images in the manuscript are provided in the source data. Correspondence and requests for materials should be addressed to F.K. Source data are provided with this paper.

References

  1. Craigie, R. & Bushman, F. D. HIV DNA integration. Cold Spring Harb. Perspect. Med. 2, a006890 (2012).

    PubMed  PubMed Central  Google Scholar 

  2. Brégnard, C., Benkirane, M. & Laguette, N. DNA damage repair machinery and HIV escape from innate immune sensing. Front. Microbiol. 5, 176 (2014).

    PubMed  PubMed Central  Google Scholar 

  3. Mamede, J. I., Cianci, G. C., Anderson, M. R. & Hope, T. J. Early cytoplasmic uncoating is associated with infectivity of HIV-1. Proc. Natl Acad. Sci. USA 114, E7169–E7178 (2017).

    CAS  PubMed  Google Scholar 

  4. Towers, G. J. & Noursadeghi, M. Interactions between HIV-1 and the cell-autonomous innate immune system. Cell Host Microbe 16, 10–18 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Yan, N., Regalado-Magdos, A. D., Stiggelbout, B., Lee-Kirsch, M. A. & Lieberman, J. The cytosolic exonuclease TREX1 inhibits the innate immune response to human immunodeficiency virus type 1. Nat. Immunol. 11, 1005–1013 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Hrecka, K. et al. HIV-1 and HIV-2 exhibit divergent interactions with HLTF and UNG2 DNA repair proteins. Proc. Natl Acad. Sci. USA 113, E3921–E3930 (2016).

    CAS  PubMed  Google Scholar 

  8. Schröfelbauer, 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).

    PubMed  PubMed Central  Google Scholar 

  9. Craigie, R., Fujiwara, T. & Bushman, F. The IN protein of Moloney murine leukemia virus processes the viral DNA ends and accomplishes their integration in vitro. Cell 62, 829–837 (1990).

    CAS  PubMed  Google Scholar 

  10. Katz, R. A., Merkel, G., Kulkosky, J., Leis, J. & Skalka, A. M. The avian retroviral IN protein is both necessary and sufficient for integrative recombination in vitro. Cell 63, 87–95 (1990).

    CAS  PubMed  Google Scholar 

  11. Daniel, R. et al. Evidence that stable retroviral transduction and cell survival following DNA integration depend on components of the nonhomologous end joining repair pathway. J. Virol. 78, 8573–8581 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Willey, R. L., Maldarelli, F., Martin, M. A. & Strebel, K. Human immunodeficiency virus type 1 Vpu protein induces rapid degradation of CD4. J. Virol. 66, 7193–7200 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Wildum, S., Schindler, M., Munch, J. & Kirchhoff, F. Contribution of Vpu, Env, and Nef to CD4 down-modulation and resistance of human immunodeficiency virus type 1-infected T cells to superinfection. J. Virol. 80, 8047–8059 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Neil, S. J. D., Zang, T. & Bieniasz, P. D. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature 451, 425–430 (2008).

    CAS  PubMed  Google Scholar 

  15. Van Damme, N. et al. The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein. Cell Host Microbe 3, 245–252 (2008).

    PubMed  PubMed Central  Google Scholar 

  16. Ryan, E. L., Hollingworth, R. & Grand, R. J. Activation of the DNA damage response by RNA viruses. Biomolecules 6, 2 (2016).

    PubMed  PubMed Central  Google Scholar 

  17. Bennardo, N., Cheng, A., Huang, N. & Stark, J. M. Alternative-NHEJ is a mechanistically distinct pathway of mammalian chromosome break repair. PLoS Genet. 4, e1000110 (2008).

  18. Akyuz, N. et al. DNA substrate dependence of p53-mediated regulation of double-strand break repair. Mol. Cell. Biol. 22, 6306–6317 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Restle, A. et al. Dissecting the role of p53 phosphorylation in homologous recombination provides new clues for gain-of-function mutants. Nucleic Acids Res. 36, 5362–5375 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Keimling, M. et al. Functional characterization connects individual patient mutations in ataxia telangiectasia mutated (ATM) with dysfunction of specific DNA double-strand break-repair signaling pathways. FASEB J. 25, 3849–3860 (2011).

    CAS  PubMed  Google Scholar 

  21. Sauter, D. et al. Differential regulation of NF-κB-mediated proviral and antiviral host gene expression by primate lentiviral Nef and Vpu proteins. Cell Rep. 10, 586–599 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Volcic, M. et al. NF-κB regulates DNA double-strand break repair in conjunction with BRCA1-CtIP complexes. Nucleic Acids Res. 40, 181–195 (2012).

    CAS  PubMed  Google Scholar 

  23. Laguette, N. et al. Premature activation of the SLX4 complex by Vpr promotes G2/M arrest and escape from innate immune sensing. Cell 156, 134–145 (2014).

    CAS  PubMed  Google Scholar 

  24. Hotter, D. et al. Primate lentiviruses use at least three alternative strategies to suppress NF-κB-mediated immune activation. PLoS Pathog. 13, e1006598 (2017).

    PubMed  PubMed Central  Google Scholar 

  25. Cooper, A. et al. HIV-1 causes CD4 cell death through DNA-dependent protein kinase during viral integration. Nature 498, 376–379 (2013).

    CAS  PubMed  Google Scholar 

  26. Kmiec, D. et al. Vpu-mediated counteraction of tetherin is a major determinant of HIV-1 interferon resistance. mBio 7, e00934-16 (2016).

    PubMed  PubMed Central  Google Scholar 

  27. Langer, S. et al. HIV-1 Vpu is a potent transcriptional suppressor of NF-κB-elicited antiviral immune responses. eLife 8, e41930 (2019).

  28. Yamada, E. et al. Human-specific adaptations in Vpu conferring anti-tetherin activity are critical for efficient early HIV-1 replication in vivo. Cell Host Microbe 23, 110–120 (2018).

    CAS  PubMed  Google Scholar 

  29. Kueck, T. & Neil, S. J. D. A cytoplasmic tail determinant in HIV-1 Vpu mediates targeting of tetherin for endosomal degradation and counteracts interferon-induced restriction. PLoS Pathog. 8, e1002609 (2012).

  30. Margottin, F. et al. A novel human WD protein, h-βTrCp, that interacts with HIV-1 Vpu connects CD4 to the ER degradation pathway through an F-box motif. Mol. Cell 1, 565–574 (1998).

    CAS  PubMed  Google Scholar 

  31. Akari, H., Bour, S., Kao, S., Adachi, A. & Strebel, K. The human immunodeficiency virus type 1 accessory protein Vpu induces apoptosis by suppressing the nuclear factor κB–dependent expression of antiapoptotic factors. J. Exp. Med. 194, 1299–1312 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Huertas, P. DNA resection in eukaryotes: deciding how to fix the break. Nat. Struct. Mol. Biol. 17, 11–16 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Maréchal, A. & Zou, L. RPA-coated single-stranded DNA as a platform for post-translational modifications in the DNA damage response. Cell Res. 25, 9–23 (2015).

    PubMed  Google Scholar 

  34. Mukherjee, B., Tomimatsu, N. & Burma, S. Immunofluorescence-based methods to monitor DNA end resection. Methods Mol. Biol. 1292, 67–75 (2015).

  35. Croteau, D. L., Popuri, V., Opresko, P. L. & Bohr, V. A. Human RecQ helicases in DNA repair, recombination, and replication. Annu. Rev. Biochem. 83, 519–552 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Nimonkar, A. V., Ozsoy, A. Z., Genschel, J., Modrich, P. & Kowalczykowski, S. C. Human exonuclease 1 and BLM helicase interact to resect DNA and initiate DNA repair. Proc. Natl Acad. Sci. USA 105, 16906–16911 (2008).

  37. Hampp, S. et al. DNA damage tolerance pathway involving DNA polymerase ι and the tumor suppressor p53 regulates DNA replication fork progression. Proc. Natl Acad. Sci. USA 113, E4311–E4319 (2016).

    CAS  PubMed  Google Scholar 

  38. Eladad, S. et al. Intra-nuclear trafficking of the BLM helicase to DNA damage-induced foci is regulated by SUMO modification. Hum. Mol. Genet. 14, 1351–1365 (2005).

    CAS  PubMed  Google Scholar 

  39. Ouyang, K. J., Yagle, M. K., Matunis, M. J. & Ellis, N. A. BLM SUMOylation regulates ssDNA accumulation at stalled replication forks. Front. Genet. 4, 167 (2013).

    PubMed  PubMed Central  Google Scholar 

  40. Dellaire, G. & Bazett-Jones, D. P. PML nuclear bodies: dynamic sensors of DNA damage and cellular stress. BioEssays 26, 963–977 (2004).

    CAS  PubMed  Google Scholar 

  41. Vancurova, M. et al. PML nuclear bodies are recruited to persistent DNA damage lesions in an RNF168-53BP1 dependent manner and contribute to DNA repair. DNA Repair 78, 114–127 (2019).

    CAS  PubMed  Google Scholar 

  42. Vigan, R. & Neil, S. J. D. Separable determinants of subcellular localization and interaction account for the inability of group O HIV-1 Vpu to counteract tetherin. J. Virol. 85, 9737–9748 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Jäger, S. et al. Global landscape of HIV–human protein complexes. Nature 481, 365–370 (2012).

    Google Scholar 

  44. Saitoh, N. et al. In situ SUMOylation analysis reveals a modulatory role of RanBP2 in the nuclear rim and PML bodies. Exp. Cell Res. 312, 1418–1430 (2006).

    CAS  PubMed  Google Scholar 

  45. Sauter, D. et al. Tetherin-driven adaptation of Vpu and Nef function and the evolution of pandemic and nonpandemic HIV-1 strains. Cell Host Microbe 6, 409–421 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Werner, A., Flotho, A. & Melchior, F. The RanBP2/RanGAP1*SUMO1/Ubc9 complex is a multisubunit SUMO E3 ligase. Mol. Cell 46, 287–298 (2012).

    CAS  PubMed  Google Scholar 

  47. Kilzer, J. M. et al. Roles of host cell factors in circularization of retroviral DNA. Virology 314, 460–467 (2003).

    CAS  PubMed  Google Scholar 

  48. Shank, P. R. et al. Mapping unintegrated avian sarcoma virus DNA: termini of linear DNA bear 300 nucleotides present once or twice in two species of circular DNA. Cell 15, 1383–1395 (1978).

    CAS  PubMed  Google Scholar 

  49. Chandramouly, G. et al. Small-molecule disruption of RAD52 rings as a mechanism for precision medicine in BRCA-deficient cancers. Chem. Biol. 22, 1491–1504 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Kan, Y., Batada, N. N. & Hendrickson, E. A. Human somatic cells deficient for RAD52 are impaired for viral integration and compromised for most aspects of homology-directed repair. DNA Repair 55, 64–75 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Lau, A., Kanaar, R., Jackson, S. P. & O’Connor, M. J. Suppression of retroviral infection by the RAD52 DNA repair protein. EMBO J. 23, 3421–3429 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Schaller, T. et al. HIV-1 capsid–cyclophilin interactions determine nuclear import pathway, integration targeting and replication efficiency. PLoS Pathog. 7, e1002439 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Di Nunzio, F. et al. Human nucleoporins promote HIV-1 docking at the nuclear pore, nuclear import and integration. PLoS ONE 7, e46037 (2012).

    PubMed  PubMed Central  Google Scholar 

  54. Zhang, R., Mehla, R. & Chauhan, A. Perturbation of host nuclear membrane component RanBP2 impairs the nuclear import of human immunodeficiency virus-1 preintegration complex (DNA). PLoS ONE 5, e15620 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Galão, R. P., Le Tortorec, A., Pickering, S., Kueck, T. & Neil, S. J. D. Innate sensing of HIV-1 assembly by tetherin induces NFκB-dependent proinflammatory responses. Cell Host Microbe 12, 633–644 (2012).

    PubMed  PubMed Central  Google Scholar 

  56. Mohammadi, P. et al. 24 hours in the life of HIV-1 in a T cell line. PLoS Pathog. 9, e1003161 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Lama, J. The physiological relevance of CD4 receptor down-modulation during HIV infection. Curr. HIV Res. 1, 167–184 (2003).

    CAS  PubMed  Google Scholar 

  58. Klimkait, T., Strebel, K., Hoggan, M. D., Martin, M. A. & Orenstein, J. M. The human immunodeficiency virus type 1-specific protein Vpu is required for efficient virus maturation and release. J. Virol. 64, 621–629 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. König, R. et al. Global analysis of host–pathogen interactions that regulate early-stage HIV-1 replication. Cell 135, 49–60 (2008).

    PubMed  PubMed Central  Google Scholar 

  60. Zhong, S. et al. A role for PML and the nuclear body in genomic stability. Oncogene 18, 7941–7947 (1999).

    CAS  PubMed  Google Scholar 

  61. Sohn, S.-Y. & Hearing, P. Adenovirus regulates sumoylation of Mre11–Rad50–Nbs1 components through a paralog-specific mechanism. J. Virol. 86, 9656–9665 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Boichuk, S., Hu, L., Makielski, K., Pandolfi, P. P. & Gjoerup, O. V. Functional connection between Rad51 and PML in homology-directed repair. PLoS ONE 6, e25814 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Lusic, M. et al. Proximity to PML nuclear bodies regulates HIV-1 latency in CD4+ T cells. Cell Host Microbe 13, 665–677 (2013).

    CAS  PubMed  Google Scholar 

  64. Erdal, E., Haider, S., Rehwinkel, J., Harris, A. L. & McHugh, P. J. A prosurvival DNA damage-induced cytoplasmic interferon response is mediated by end resection factors and is limited by Trex1. Genes Dev. 31, 353–369 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Himmels, S.-F. & Sartori, A. A. Controlling DNA-end resection: an emerging task for ubiquitin and SUMO. Front. Genet. 7, 152 (2016).

    PubMed  PubMed Central  Google Scholar 

  66. González-Prieto, R., Cuijpers, S. A. G., Luijsterburg, M. S., van Attikum, H. & Vertegaal, A. C. O. SUMOylation and PARylation cooperate to recruit and stabilize SLX4 at DNA damage sites. EMBO Rep. 16, 512–519 (2015).

    PubMed  PubMed Central  Google Scholar 

  67. Kirby, T. J. & Lammerding, J. Emerging views of the nucleus as a cellular mechanosensor. Nat. Cell Biol. 20, 373–381 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Vermeire, J. et al. HIV triggers a cGAS-dependent, Vpu- and Vpr-regulated type I interferon response in CD4+ T cells. Cell Rep. 17, 413–424 (2016).

    CAS  PubMed  Google Scholar 

  69. Levy, J. A., Virolainen, M. & Defendi, V. Human lymphoblastoid lines from lymph node and spleen. Cancer 22, 517–524 (1968).

    CAS  PubMed  Google Scholar 

  70. Pedrazzi, G. Direct association of Bloom’s syndrome gene product with the human mismatch repair protein MLH1. Nucleic Acids Res. 29, 4378–4386 (2001).

  71. Uhl, M. et al. Role of SIRT1 in homologous recombination. DNA Repair 9, 383–393 (2010).

    CAS  PubMed  Google Scholar 

  72. Gole, B., Baumann, C., Mian, E., Ireno, C. I. & Wiesmüller, L. Endonuclease G initiates DNA rearrangements at the MLL breakpoint cluster upon replication stress. Oncogene 34, 3391–3401 (2015).

    CAS  PubMed  Google Scholar 

  73. Obermeier, K. et al. Heterozygous PALB2 c.1592delT mutation channels DNA double-strand break repair into error-prone pathways in breast cancer patients. Oncogene 35, 3796–3806 (2016).

    CAS  PubMed  Google Scholar 

  74. Lambert, S. & Lopez, B. S. Characterization of mammalian RAD51 double strand break repair using non-lethal dominant-negative forms. EMBO J. 19, 3090–3099 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Sauter, D. et al. Human tetherin exerts strong selection pressure on the HIV-1 group N Vpu protein. PLoS Pathog. 8, e1003093 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Herzer, K., Weyer, S., Krammer, P. H., Galle, P. R. & Hofmann, T. G. Hepatitis C virus core protein inhibits tumor suppressor protein promyelocytic leukemia function in human hepatoma cells. Cancer Res. 65, 10830–10837 (2005).

  77. Collis, S. J., Swartz, M. J., Nelson, W. G. & DeWeese, T. L. Enhanced radiation and chemotherapy-mediated cell killing of human cancer cells by small inhibitory RNA silencing of DNA repair factors. Cancer Res. 63, 1550–1554 (2003).

    CAS  PubMed  Google Scholar 

  78. Lallemand-Breitenbach, V. et al. Arsenic degrades PML or PML–RARα through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat. Cell Biol. 10, 547–555 (2008).

    CAS  PubMed  Google Scholar 

  79. Jacque, J.-M. & Stevenson, M. The inner-nuclear-envelope protein emerin regulates HIV-1 infectivity. Nature 441, 641–645 (2006).

    CAS  PubMed  Google Scholar 

  80. Rao, X., Huang, X., Zhou, Z. & Lin, X. An improvement of the 2ΔΔCT method for quantitative real-time polymerase chain reaction data analysis. Biostat. Bioinforma. Biomath. 3, 71–85 (2013).

    PubMed  PubMed Central  Google Scholar 

  81. Munir, S., Thierry, S., Subra, F., Deprez, E. & Delelis, O. Quantitative analysis of the time-course of viral DNA forms during the HIV-1 life cycle. Retrovirology 10, 87 (2013).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Schloesser for technical help with the PLA experiments, V. Lallemand-Breitenbach for providing us with the antibody recognizing PML isoforms I–IV, M. Lusic for support and helpful discussions, and J. M. Stark for giving us the EJ5SceGFP construct for NHEJ measurements. The Quadro P6000 used for this research project was made available by the NVIDIA Corporation. L.W. was supported by grants from the German Research Foundation (grant no. DFG WI 3099/7, B05 in CRC 1279), European–German Space Agency (ESA/DLR) and German Ministry of Economy (BMWi, A0-10-IBER-2 funding grant no. 50WB1225). F.K. was supported by the DFG (grant no. KI 548/11-1; CRC 1279) and an Advanced ERC investigator grant (Anti-Virome). D.S., K.M.J.S. and F.K. are funded by the DFG priority program grant no. SPP1923 and T.G.H. is supported by CRC 1361. K.M.J.S. was supported by a Marie Skłodowska-Curie Individual Fellowship from the European Union’s Framework Program for Research and Innovation Horizon 2020 (2014–2020) under grant agreement no. 794803 (‘VIAR’) and the DFG (grant no. SP1600/4-1).

Author information

Authors and Affiliations

Authors

Contributions

M.V. performed and analysed most of the experiments. K.M.J.S., L.K., D.H. and D.S. contributed to additional assay work, image analysis and experimental expertise. C.M.S. performed the cloning. M.S. and T.S. contributed to the assays to analyse PML. N.J.A. contributed expertise and T.G.H. contributed materials and expertise. M.V., K.M.J.S., L.W. and F.K. wrote the manuscript. M.V., L.W. and F.K. supervised the study.

Corresponding authors

Correspondence to Lisa Wiesmüller or Frank Kirchhoff.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data

Extended Data Fig. 1 Primary DSB repair data and controls.

Primary DSB repair data and Role of DNA damage sensors. a, Primary FACS data for EGFP-based evaluation of DSB repair as in Fig. 1d. Evaluation of DSB repair frequency described in Methods section. b-c, Apoptosis induction and cell cycle distribution in Jurkat cells transfected with pHIV-1-NL4-3-env*-IRES-mCherry (HIV-1), pCMV-HA-I-SceI plus repair reporter plasmid HR-EGFP/5´EGFP. In parallel, split samples were treated with caspase inhibitor zVAD-fmk. Cells were fixed 48 h post-transfection with ethanol/acetone and stained with propidium iodide for DNA content analysis. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, *p=0.0313. In c, samples in the same cell cycle phase were statistically analyzed. d, Extrachromosomal homologous repair frequency (Hom. repair freq.) in WTK1 cells transfected with pHIV-1-NL4-3-env*-IRES-mCherry, pCMV-HA-I-SceI plus repair plasmid HR-EGFP/3´EGFP for detection of HR and SSA (construct on top). Split samples were treated with zVAD-fmk. Bars represent means ± SEM, n=3 biologically independent experiments in triplicates, *p=0.0391. Control samples transfected with control plasmid set to 100 % (absolute mean value: 2 %). e-f, Apoptosis induction and cell cycle distribution in WTK1(HR/3′) cells 48 h after transfection with HIV-1 plus pCMV-HA-I-SceI. Bars represent means ± SEM, n=2 biologically independent experiments in duplicates (f) or triplicates (g). In (f) samples in the same cell cycle phase were statistically analyzed. g, Intrachromosomal homologous repair frequency in KMV(Δ/3′) cells transfected with HIV-1 plus pCMV-HA-I-SceI. Split samples were treated with and without Hygromycin B (60 µg/ml) 4 h post-transfection. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, *p=0.0313, (absolute mean value: 0.2 %). Note the schematic presentation of the chromosomally integrated repair construct ΔEGFP/3´EGFP in KMV(Δ/3′) with a Hygromycin resistance cassette (blue bar) on top. h-l, Homologous repair frequency in WTK1(HR/3′) cells co-transfected with pHIV-1-NL4-3-env*-IRES-mCherry, pCMV-HA-I-SceI and shRNA to downregulate ATM (a), ATR (b), Nibrin (c), CtIP (d) and Ku70 (e) and corresponding empty vectors in the controls. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, *p=0.0313 (absolute mean value: 0.04%). Lower panels show Western blots demonstrating efficiency of knockdowns. Two sided Wilcoxon matched-pairs test in b, d, g, h-l.

Source data

Extended Data Fig. 2 Vpu expression in different experimental settings and controls to exclude potentially confounding factors.

Vpu expression in different experimental settings and controls to exclude potentially confounding factors. a, Vpu titration. Evaluation of DSB repair frequencies in WTK1(HR/3′) cells transfected with increasing amounts of NL4-3 Vpu-AU1 expression plasmid. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, *p=0.0313. Lower panels show Western blot demonstrating expression efficiency. b, Expression of WITO Vpu-AU1 in WTK1(HR/3′) cells transduced with pHIV-1-NL4-3 WITO Vpu-AU1 compared to cells transfected with pCG-WITO Vpu-AU1 expression vector or the pHIV-1-NL4-3 WITO Vpu-AU1 proviral construct. In the proviral construct vpu and env were separated and the NL-4-3 vpu replaced by an AU-1-tagged WITO vpu. 24 h post-transfection samples were analyzed by Western blot. c, Apoptosis and cell cycle analysis in WTK1(HR/3′) cells after transfection with pCG-NL4-3 Vpu-AU1 expression plasmid. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, statistical analysis was calculated for comparisons between different samples in the same cell cycle phase. d, Infection rates. Graph presenting mean values ± SD of p24 stained in CD4+ T cells from 3 donors shown in Fig. 3d. Right, primary flow cytometry data. Percentages of p24 positive cells in the live cell population (SSC/FSC gate) were calculated. e, Homologous repair, apoptosis and cell cycle in primary HIV-1 transfected CD4+ T cells. Cells were transfected with infectious (env+) HIV-1 CH058 wt, vpr* or vpu* constructs, together with repair reporter and meganuclease I-SceI expression plasmid. 48 h post-transfection samples were examined for repair frequencies or aceton/ethanol fixed and propidium iodide stained to evaluate cell cycle and apoptosis. Bars represent means ± SEM, n=3 biologically independent experiments in triplicates, *p=0.0152, **p=0.0022 (absolute mean value of homologous repair frequency was 0.3%). Two sided Wilcoxon matched-pairs test in a and e.

Source data

Extended Data Fig. 3 Analysis of mutant Vpu proteins and evaluation of known cellular Vpu targets.

Analysis of mutant Vpu proteins and evaluation of known cellular Vpu targets. a, Sequence of WITO Vpu with mutated positions indicated. b, Western blot depicting expression of different WITO Vpu mutants from (a) and CH106 wt and its KKDQ mutant with the ER retention sequence. Experiment repeated 2 times with similar outcome. c, Homologous repair in WTK1(HR/3′) cells transfected with different mutants of WITO Vpu or CH106 and the CH106 KKDQ mutant with ER retention sequence. 48 h post transfection cells were analyzed by FACS to evaluate DSB repair frequencies. Bars represent means ± SEM, n=3 or 4 biologically independent experiments in triplicates, *p=0.0273, **p=0.0078, **p=0.0039 (absolute mean value: 0.7 %). Statistical evaluation shown for the comparison between wt WITO Vpu and the indicated Ala substitution mutants. d, BrdU assay to analyze DNA processing mediated by different WITO Vpu mutants. WTK1(HR/3′) WITO transfected cells were IR (2 Gy), pre-extracted, fixed and stained against BrdU. Data presents 4 independent experiments where each dot (n=14-18) presents mean value of 30-50 analyzed nuclei from one image view, ± SEM. *p=0.0166, **p=0.0012, **p=0.0085, ****p=0.0001. Black stars presenting comparison to the control, blue stars comparison to WITO Vpu. The lower panel shows representative BrdU and AU1 microscopic images. e, Homologous repair in WTK1(HR/3′) (β-TrCP). Cells were transfected with wt, vpr* or vpu* pHIV-1-NL4-3-env*-IRES-mCherry constructs together with expression plasmids for I-SceI and the DN form of β-TrCP. 48 h later cells were harvested for FACS analysis. Bars represent means ± SEM, n=3 biologically independent experiments in triplicates, **p=0.039 (absolute mean value: 0.1%). Lower panels show Western blot demonstrating overexpression of β-TrCP-DN. f, Immunoprecipitation (IP) of RAD52 1h after γ-irradiation (2Gy) of WTK1(HR/3′) cells transfected 24h before with NL4-3 Vpu expression plasmid or empty vector (Control). Graph presents mean values ± SD from two independent experiments. Note that RAD52 is difficult to detect by Western blotting unless it is concentrated for example by IP. Thus, most analyses had to rely on mRNA expression levels. g, Evaluation of RAD52 mRNA levels in SaOS cells treated as described in the legend Fig. 2i. Bars represent means ± SEM, n=4. Two sided Wilcoxon matched-pairs test in c, d, e, g.

Source data

Extended Data Fig. 4 Role of 53BP1 and cellular nucleases in HIV-1-mediated homologous repair regulation.

Role of 53BP1 and cellular nucleases in HIV-1-mediated homologous repair regulation. a, Detection of DSBs by 53BP1 foci. WTK1(HR/3′) cells transfected with wt or vpu* pHIV-1-NL4-3-env*-IRES-mCherry constructs were γ-irradiated (2Gy) or left untreated (0Gy), fixed and stained for 53BP1 at the indicated time points. Immunolabeled foci were scored by quantification in 150 nuclei each, n=3 biologically independent experiments. Maximum scores were set to 100% (absolute foci numbers for 53BP1: 7), bars represent means ±SD. The left panel shows representative microscopic images of 53BP1 foci. b, Role of DNase2, ERCC1 and FEN1 nucleases in HIV-1 mediated DNA repair. WTK1(HR/3′) cells were transfected with pHIV-1-NL4-3-env*-IRES-mCherry, I-SceI-HA and shRNA to silence DNase2, ERCC1 or FEN1. Bars represent means ± SEM, n=2 (DNase2, ERCC1) or n=4 (FEN1) biologically independent experiments in triplicates, *p=0.0313 (absolute mean values: Dnase2 2.2%, ERCC1 0.3% and FEN1 1.0%). Right three graphs presenting rtPCR analysis to show DNase2, ERCC1 and FEN1 knockdown efficiencies. Bars represent means ± SEM, n=4 biologically independent experiments, two sided unpaired t-test, ***p=0.0001. c, Homologous repair frequency in WTK1(HR/3′) cells 48 h after transfection with pHIV-1-NL4-3-env*-IRES-mCherry constructs containing intact or defective vpu genes, pCMV-HA-I-SceI and shRNA to silence SLX4. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, *p=0.0313 (absolute mean value: 0.1%). d, Western blot analysis of WTK1(HR/3′) cells 48h after transfection with pHIV-1-NL4-3-env*-IRES-mCherry and pCMV-HA-I-SceI plasmids. The left panel shows examples of primary data and the right panel mean values ± SD of 3-6 independent experiments after quantification of band intensities using Bio-Rad ChemiDoc. Two sided Wilcoxon matched-pairs test in b (left three panels) and c.

Source data

Extended Data Fig. 5 Role of BLM and EXO1 in Vpu-mediated modulation of DNA end processing.

Role of BLM and EXO1 in Vpu-mediated modulation of DNA end processing. a, Representative images for immunofluorescence analysis of p-RPA and BrdU and b, knock-down efficiency of BLM and EXO1. c, p-RPA accumulation as a function of BLM. WTK1(HR/3′) cells were γ-irradiated (2Gy) 48h after transfection with wt or vpu* pHIV-1-NL4-3-env*-IRES-mCherry plus shRNA to silence BLM (BLMkd). 1h post-irradiation, samples were taken for p-RPA32 S4/8 and RPA Western Blot analysis. The right panel shows mean values (±SD) of p-RPA levels obtained in three independent experiments relative to control cells. d, p-RPA Western blot analysis in WTK1(HR/3′) cells transfected with wt or vpu* pHIV-1-NL4-3-env*-IRES-mCherry, together with shRNA against EXO1. 48 h after transfection cells were irradiated and taken for Western blotting. e, Expression of BLM and EXO1 in primary CD4+ T cells. CD4+ T cells isolated form two blood donors were either treated with IL-2 alone or stimulated with IL-2/PHA or IL-2 and CD3/CD28 magnetic beads or left untreated (U) for 3d when cells were harvested for Western blot analysis.

Source data

Extended Data Fig. 6 Effect of Vpu on PML-NBs. Role of BLM and EXO1 in Vpu-medidated modulation of DNA end processing.

Effect of Vpu on PML-NBs. Role of BLM and EXO1 in Vpu-mediated modulation of DNA end processing. a, Immunofluorescence analysis of Sp100 foci formation in WTK1(HR/3′) cells transfected with 1µg NL4-3, WITO Vpu or empty control. Cells were fixed at indicated time points. Data presents 3 independent experiments where each dot (n=6) represents mean value of 30-50 analyzed cells from one microscopic view; bars represent means ±SEM, *p=0.0313. Right panel, representative microscopic images. b, c, Immunofluorescence analysis of PML-NB and Sp100 foci in WTK1(HR/3′) cells transfected with wt or vpu* pHIV-1-CH058-env*-IRES-BFP constructs. Data present three independent experiments where each dot represents mean value of 30-50 analyzed cells from one microscopic view; bars represent means ±SEM, *p=0.0313. Right panel, representative microscopic images. Two sided Wilcoxon matched-pairs test in a-c.

Extended Data Fig. 7 Localization of Vpu at the nuclear membrane and effects on RanBP2-RanGAP1 proximity and function.

Localization of Vpu at the nuclear membrane and effects on RanBP2-RanGAP1 proximity and function. a, Subcellular protein fractionation performed in CD4+ T cells transfected with pCG-WITO Vpu-AU1 expression plasmid or empty control. Experiment conducted 2 times with similar outcome. b, Confocal microscopic localization analysis of Vpu and RanBP2. SaOS cells were transfected with 1 µg SIVcpz MB897 Vpu-AU1. 24 h later cells were fixed, permeabilized and costained against AU1-tagged Vpu and RanBP2. Graph on left presents the probability of Vpu following RanBP2 calculated from the images such as displayed on the right, bars represent means ± SEM, Two sided unpaired t- test here and in c, n=6, ****p=0.0001. Size bar = 5 µm. c, Primary CD4+ T cells were transfected with 0.5 µg of a vector expressing MB897 Vpu-AU1. Cells were permeabilized, stained with anti-AU1 and RanBP2 Ab and analyzed by confocal microscopy 24h post-transfection. Graph on left presents the probability of Vpu following RanBP2 calculated from the images such as displayed on the right, bars represent means ±SEM, n=5, ****p=0.0001. Size bar =5 µm. d, RanBP2 and RanGAP1 PLA in primary CD4+ T cells. Cells were transfected with 0.5 µg WITO Vpu-AU1 expression plasmid or empty vector control. Left, evaluation of RanBP2-RanGAP1 PLA foci per cell from two different donors, n=32, bars represent means ± SEM, two sided Mann-Whitney test, ****p=0.0001. Right, representative microscopic images. e, PLA to evaluate RanGAP1 and SUMO1 interaction in WTK1(HR/3′) cells transfected with WITO Vpu-AU1 expression plasmid or empty control. Left panel presenting evaluation of RanGAP1-SUMO PLA foci per cell, n=4 biologically independent experiments; two sided Wilcoxon matched-pairs test, mean values ± SEM, right panel presenting microscopic images. f, PLA assay to evaluate EXO1 and SUMO2/3 interaction in WTK1(HR/3′) cells transfected with WITO Vpu-AU1 expression plasmid or empty control. Left panel presenting evaluation of EXO1-SUMO2/3 foci per cell, n=2 biologically independent experiments, two sided Wilcoxon matched-pairs test (control n= 27, WITO n=20), mean values ± SEM, ****p=0.0001. Right panel presenting microscopic images.

Source data

Extended Data Fig. 8 Role of PIAS1, PIAS4 and RanBP2 SUMO ligases in BLM SUMOylation.

Role of PIAS1, PIAS4 and RanBP2 SUMO ligases in BLM SUMOylation. a, Role of PIAS1, PIAS4 and RanBP2 SUMO ligases in BLM SUMOylation. WTK1(HR/3′) cells were transfected with 1 µg WITO Vpu-AU1 or control plus 3 µg of shRNA to target PIAS1, PIAS4 and RanBP2. 24h later PLA was performed. Left, evaluation of BLM-SUMO2/3 foci per cell, n=3 biologically independent experiments where each dot (8-12) represents a mean from 30-50 cells analyzed from one microscopic view; bars represent means ± SEM, two sided Wilcoxon matched-pairs test, *p=0.0117, **p=0.002, **p=0.0039, **p=0.0078. Right, Western blots presenting knockdown of PIAS1, PIAS4 and RanBP2. Lower panels, representative microscopic images. b, WTK1(HR/3′) cells were transfected with 1 µg WITO Vpu-AU1 or control and stained against SUMO1, RanBP2 and RanGAP1 24h later. Intensity profiles including the corresponding DAPI (blue) staining in the presence (red) or absence (black) of WITO vpu were quantified using ImageJ. Displayed are the means (full lines) of 10µm profiles of 25 individually quantified and aligned cells including SEM (dotted line). Exemplary confocal images are shown next to the quantifications and the profile indicated by a white line.

Source data

Extended Data Fig. 9 Effect of Vpu and RAD52 or BLM on unintegrated viral cDNA, integration and superinfection in primary CD4+ T cells.

Effect of Vpu and RAD52 or BLM on unintegrated viral cDNA, integration and superinfection in primary CD4+ T cells. a-c, rtPCR analysis of 1-LTR, 2-LTR circles and integration in CD4+ T cells infected with wt or vpu* pHIV-1-NL4-3 in the presence or absence of a RAD52 inhibitor (5 µM 6-Hydroxy-DL-DOPA) 48 h post infection. Bars represent means ± SEM, n=3 biologically independent experiments in triplicates, (a) *p=0.0156, **p=0.0039, (b) **p=0.0078, (c) *p=0.0273, **p=0.0039. d, Propidium iodide staining of acetone/ethanol fixed cells and p24 stain of primary CD4+ T cells treated as in Fig. 7 (ac). n=3, bars represent means ± SEM of 6 (apoptosis) or 4 measurements, **p=0.0022. e, Western blot analysis of p24 protein expression in CD4+ T cells infected with wt or vpu* pHIV-1-NL4-3. Experiment conducted two times with similar outcome. f-k, rtPCR analysis of 1-LTR, 2-LTR circles and integration in CD4+ T cells transduced with wt, vpu* or vpr* defective pHIV-1-NL4-3-env* constructs (f-h) or infected with replication-competent pHIV-1-NL4-3 in the presence of the protease inhibitor saquinavir (i-k). Bars represent means ± SEM from 6 (f,g), 5 (h), 3(i), 2(j, k) donors are shown. Analysis done 48 h post-infection/transduction. l, Western blot demonstrating BLM knockdown efficiency in Jurkat cells. Experiment conducted 2 times with similar outcome. m-o, rtPCR analysis of 1-LTR and 2-LTR circles and integration in BLM silenced Jurkat cells infected with wt or vpu* VSV-G pseudotyped pHIV-1-NL4-3. Bars represent means ± SEM, n=3 biologically independent experiments in triplicates, (m) *p=0.0156, **p=0.0039; (n) *p=0.0313, **p=0.0039, (o) **p=0.0039. Analysis done 48 h post-infection. p, Supernatants from m-o were taken to analyze infectious yields determined by β-galactosidase assay in TZM-bl cells. Bars represent means ± SEM, n=2 biologically independent experiments in triplicates, *p=0.0313. Two sided Wilcoxon matched-pairs test in a-d, m-p.

Source data

Extended Data Fig. 10 Effect of Vpu on HIV-1 superinfection in primary CD4+ T cells and BLM deficient cells.

Effect of Vpu on HIV-1 superinfection in primary CD4+ T cells and BLM deficient cells. a, Schematic presentation of the experimental design to measure and calculate HIV-1 superinfection. b, Primary flow cytometry data of CD4+ T cells treated as described in panel b; n=6. Calculated percentages of superinfected cells are shown between the dot plot panels. c, Primary flow cytometry data of BLM+/- cells treated as described in the legend to Fig. 6r.

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Supplementary Table 1.

Reporting Summary

Supplementary Video 1

Video presenting colocalization between WITO Vpu and RanBP2 in transfected SaOS cells.

Supplementary Video 2

Video presenting colocalization between SIV Vpu and RanBP2 in transfected CD4+ T cells.

Supplementary Video 3

Video presenting colocalization between SIV Vpu and RanBP2 in transfected SaOS cells.

Source data

Source Data Fig. 1

Unprocessed western blots and gel.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Volcic, M., Sparrer, K.M.J., Koepke, L. et al. Vpu modulates DNA repair to suppress innate sensing and hyper-integration of HIV-1. Nat Microbiol 5, 1247–1261 (2020). https://doi.org/10.1038/s41564-020-0753-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-0753-6

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