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.

  • Review Article
  • Published:

Structure and function of retroviral integrase

Abstract

A hallmark of retroviral replication is establishment of the proviral state, wherein a DNA copy of the viral RNA genome is stably incorporated into a host cell chromosome. Integrase is the viral enzyme responsible for the catalytic steps involved in this process, and integrase strand transfer inhibitors are widely used to treat people living with HIV. Over the past decade, a series of X-ray crystallography and cryogenic electron microscopy studies have revealed the structural basis of retroviral DNA integration. A variable number of integrase molecules congregate on viral DNA ends to assemble a conserved intasome core machine that facilitates integration. The structures additionally informed on the modes of integrase inhibitor action and the means by which HIV acquires drug resistance. Recent years have witnessed the development of allosteric integrase inhibitors, a highly promising class of small molecules that antagonize viral morphogenesis. In this Review, we explore recent insights into the organization and mechanism of the retroviral integration machinery and highlight open questions as well as new directions in the field.

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: The retroviral integration process.
Fig. 2: Prototype foamy virus intasome structures and integrase active site mechanics.
Fig. 3: Diversity of retroviral intasome architectures.
Fig. 4: Integration into nucleosomal DNA.
Fig. 5: Integrase strand transfer inhibitors.
Fig. 6: Allosteric HIV-1 integrase inhibitors.

Similar content being viewed by others

References

  1. Poiesz, B. J. et al. Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc. Natl Acad. Sci. USA 77, 7415–7419 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Coffin, J. M. The discovery of HTLV-1, the first pathogenic human retrovirus. Proc. Natl Acad. Sci. USA 112, 15525–15529 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Barre-Sinoussi, F. et al. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 220, 868–871 (1983).

    CAS  PubMed  Google Scholar 

  4. Gallo, R. C. et al. Isolation of human T-cell leukemia virus in acquired immune deficiency syndrome (AIDS). Science 220, 865–867 (1983).

    CAS  PubMed  Google Scholar 

  5. Baltimore, D. RNA-dependent DNA polymerase in virions of RNA tumour viruses. Nature 226, 1209–1211 (1970).

    CAS  PubMed  Google Scholar 

  6. Temin, H. M. & Mizutani, S. RNA-dependent DNA polymerase in virions of Rous sarcoma virus. Nature 226, 1211–1213 (1970).

    CAS  PubMed  Google Scholar 

  7. Schiff, R. D. & Grandgenett, D. P. Virus-coded origin of a 32,000-dalton protein from avian retrovirus cores: structural relatedness of p32 and the beta polypeptide of the avian retrovirus DNA polymerase. J. Virol. 28, 279–291 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Jacks, T. et al. Characterization of ribosomal frameshifting in HIV-1 gag-pol expression. Nature 331, 280–283 (1988).

    CAS  PubMed  Google Scholar 

  9. Carlson, L. A. et al. Three-dimensional analysis of budding sites and released virus suggests a revised model for HIV-1 morphogenesis. Cell Host Microbe 4, 592–599 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen, N. Y. et al. HIV-1 capsid is involved in post-nuclear entry steps. Retrovirology 13, 28 (2016).

    PubMed  PubMed Central  Google Scholar 

  11. Burdick, R. C. et al. HIV-1 uncoats in the nucleus near sites of integration. Proc. Natl Acad. Sci. USA 117, 5486–5493 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Zila, V. et al. Cone-shaped HIV-1 capsids are transported through intact nuclear pores. Cell 184, 1032–1046.e18 (2021). This study provides visible evidence that intact viral cores pass through cellular nuclear pore complexes during HIV-1 nuclear entry.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Li, C., Burdick, R. C., Nagashima, K., Hu, W. S. & Pathak, V. K. HIV-1 cores retain their integrity until minutes before uncoating in the nucleus. Proc. Natl Acad. Sci. USA 118, e2019467118 (2021). Together with Burdick et al. (2020), imaging of HIV-1 infection reveals that viral cores remain largely intact until uncoating in the nucleus near sites of integration.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Lee, M. S. & Craigie, R. A previously unidentified host protein protects retroviral DNA from autointegration. Proc. Natl Acad. Sci. USA 95, 1528–1533 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Lin, C. W. & Engelman, A. The barrier-to-autointegration factor is a component of functional human immunodeficiency virus type 1 preintegration complexes. J. Virol. 77, 5030–5036 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Farnet, C. M. & Bushman, F. D. HIV-1 cDNA integration: requirement of HMG I(Y) protein for function of preintegration complexes in vitro. Cell 88, 483–492 (1997).

    CAS  PubMed  Google Scholar 

  17. Llano, M. et al. LEDGF/p75 determines cellular trafficking of diverse lentiviral but not murine oncoretroviral integrase proteins and is a component of functional lentiviral preintegration complexes. J. Virol. 78, 9524–9537 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Machida, S. et al. Exploring histone loading on HIV DNA reveals a dynamic nucleosome positioning between unintegrated and integrated viral genome. Proc. Natl Acad. Sci. USA 117, 6822–6830 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Winans, S. & Goff, S. P. Mutations altering acetylated residues in the CTD of HIV-1 integrase cause defects in proviral transcription at early times after integration of viral DNA. PLoS Pathog. 16, e1009147 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Yoder, K. E. & Bushman, F. D. Repair of gaps in retroviral DNA integration intermediates. J. Virol. 74, 11191–11200 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Knyazhanskaya, E. et al. NHEJ pathway is involved in post-integrational DNA repair due to Ku70 binding to HIV-1 integrase. Retrovirology 16, 30 (2019).

    PubMed  PubMed Central  Google Scholar 

  22. Cai, M. et al. Solution structure of the N-terminal zinc binding domain of HIV-1 integrase. Nat. Struct. Biol. 4, 567–577 (1997).

    CAS  PubMed  Google Scholar 

  23. Dyda, F. et al. Crystal structure of the catalytic domain of HIV-1 integrase: similarity to other polynucleotidyl transferases. Science 266, 1981–1986 (1994). The first high-resolution structure of a retroviral IN CCD reveals similarity to other polynucleotidyltransferases, including DNA transposases and RNase H.

    CAS  PubMed  Google Scholar 

  24. Eijkelenboom, A. P. et al. The DNA-binding domain of HIV-1 integrase has an SH3-like fold. Nat. Struct. Biol. 2, 807–810 (1995).

    CAS  PubMed  Google Scholar 

  25. Hare, S., Gupta, S. S., Valkov, E., Engelman, A. & Cherepanov, P. Retroviral intasome assembly and inhibition of DNA strand transfer. Nature 464, 232–236 (2010). The first high-resolution structure of a retroviral intasome reveals the IN–viral DNA architecture underlying DNA recombination as well as the mechanism of clinical INSTI action.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Guan, R. et al. X-ray crystal structure of the N-terminal region of Moloney murine leukemia virus integrase and its implications for viral DNA recognition. Proteins 85, 647–656 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Harshey, R. M. The Mu story: how a maverick phage moved the field forward. Mob. DNA 3, 21 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Hare, S., Maertens, G. N. & Cherepanov, P. 3′-processing and strand transfer catalysed by retroviral integrase in crystallo. EMBO J. 31, 3020–3028 (2012). This study visualizes the 3′-processing and strand transfer reactions in crystals of PFV intasomes, which defined the roles of chemical nucleophiles and divalent metal ions in catalysis, and reveals substrate mimicry by INSTIs.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Maertens, G. N., Hare, S. & Cherepanov, P. The mechanism of retroviral integration from X-ray structures of its key intermediates. Nature 468, 326–329 (2010). The first high-resolution structures of retroviral target capture and strand transfer intasome complexes reveal the requirement for target DNA distortion during integration.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Yin, Z. et al. Crystal structure of the Rous sarcoma virus intasome. Nature 530, 362–366 (2016). Together with Ballandras-Colas et al. (2016), these intasome structures first reveal the requirement for more than four IN protomers for functional integration.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ballandras-Colas, A. et al. Cryo-EM reveals a novel octameric integrase structure for betaretroviral intasome function. Nature 530, 358–361 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Ballandras-Colas, A. et al. A supramolecular assembly mediates lentiviral DNA integration. Science 355, 93–95 (2017). Cryo-EM structures of intasomes derived from the sheep lentivirus MVV reveal a higher-order macromolecular machine composed of 16 IN protomers and define the conserved intasome core shared by all retroviral intasome structures.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Passos, D. O. et al. Cryo-EM structures and atomic model of the HIV-1 strand transfer complex intasome. Science 355, 89–92 (2017). Initial cryo-EM structures of HIV-1 strand transfer complex intasomes reveal a higher-order IN dodecamer architecture.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, M. et al. A peptide derived from lens epithelium-derived growth factor stimulates HIV-1 DNA integration and facilitates intasome structural studies. J. Mol. Biol. 432, 2055–2066 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Cook, N. J. et al. Structural basis of second-generation HIV integrase inhibitor action and viral resistance. Science 367, 806 (2020). High-resolution cryo-EM structures of simian immunodeficiency virus intasomes reveal the mechanism of INSTI resistance.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Passos, D. O. et al. Structural basis for strand-transfer inhibitor binding to HIV intasomes. Science 367, 810 (2020). This study provides high-resolution cryo-EM structures of HIV-1 intasomes with several advanced INSTIs.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Bhatt, V. et al. Structural basis of host protein hijacking in human T-cell leukemia virus integration. Nat. Commun. 11, 3121 (2020). Together with Barski et al. (2020), cryo-EM studies reveal that deltaretrovirus intasomes are composed of IN tetramers and visualize details of the IN–PP2A interface.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Barski, M. S. et al. Cryo-EM structure of the deltaretroviral intasome in complex with the PP2A regulatory subunit B56γ. Nat. Commun. 11, 5043 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Pandey, K. K. et al. Cryo-EM structure of the Rous sarcoma virus octameric cleaved synaptic complex intasome. Commun. Biol. 4, 330 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. McCord, M. et al. Purification of recombinant Rous sarcoma virus integrase possessing physical and catalytic properties similar to virion-derived integrase. Protein Expr. Purif. 14, 167–177 (1998).

    CAS  PubMed  Google Scholar 

  41. Ballandras-Colas, A., Naraharisetty, H., Li, X., Serrao, E. & Engelman, A. Biochemical characterization of novel retroviral integrase proteins. PLoS ONE 8, e76638 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hare, S. et al. Structural basis for functional tetramerization of lentiviral integrase. PLoS Pathog. 5, e1000515 (2009).

    PubMed  PubMed Central  Google Scholar 

  43. Cherepanov, P. et al. HIV-1 integrase forms stable tetramers and associates with LEDGF/p75 protein in human cells. J. Biol. Chem. 278, 372–381 (2003).

    CAS  PubMed  Google Scholar 

  44. Lodi, P. J. et al. Solution structure of the DNA binding domain of HIV-1 integrase. Biochemistry 34, 9826–9833 (1995).

    CAS  PubMed  Google Scholar 

  45. Wei, S. Q., Mizuuchi, K. & Craigie, R. A large nucleoprotein assembly at the ends of the viral DNA mediates retroviral DNA integration. EMBO J. 16, 7511–7520 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, H., Wei, S. Q. & Engelman, A. Multiple integrase functions are required to form the native structure of the human immunodeficiency virus type I intasome. J. Biol. Chem. 274, 17358–17364 (1999).

    CAS  PubMed  Google Scholar 

  47. Yang, W., Lee, J. Y. & Nowotny, M. Making and breaking nucleic acids: two-Mg2+-ion catalysis and substrate specificity. Mol. Cell 22, 5–13 (2006).

    CAS  PubMed  Google Scholar 

  48. Nowotny, M. & Yang, W. Stepwise analyses of metal ions in RNase H catalysis from substrate destabilization to product release. EMBO J. 25, 1924–1933 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Engelman, A., Mizuuchi, K. & Craigie, R. HIV-1 DNA integration: mechanism of viral DNA cleavage and DNA strand transfer. Cell 67, 1211–1221 (1991).

    CAS  PubMed  Google Scholar 

  50. Chow, S. A. & Brown, P. O. Juxtaposition of two viral DNA ends in a bimolecular disintegration reaction mediated by multimers of human immunodeficiency virus type 1 or murine leukemia virus integrase. J. Virol. 68, 7869–7878 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Richardson, J. M., Colloms, S. D., Finnegan, D. J. & Walkinshaw, M. D. Molecular architecture of the Mos1 paired-end complex: the structural basis of DNA transposition in a eukaryote. Cell 138, 1096–1108 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Montano, S. P., Pigli, Y. Z. & Rice, P. A. The mu transpososome structure sheds light on DDE recombinase evolution. Nature 491, 413–417 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Morris, E. R., Grey, H., McKenzie, G., Jones, A. C. & Richardson, J. M. A bend, flip and trap mechanism for transposon integration. eLife 5, e15537 (2016).

    PubMed  PubMed Central  Google Scholar 

  54. Ghanim, G. E., Kellogg, E. H., Nogales, E. & Rio, D. C. Structure of a P element transposase-DNA complex reveals unusual DNA structures and GTP-DNA contacts. Nat. Struct. Mol. Biol. 26, 1013–1022 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Aiyer, S. et al. Structural and sequencing analysis of local target DNA recognition by MLV integrase. Nucleic Acids Res. 43, 5647–5663 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Serrao, E. et al. Integrase residues that determine nucleotide preferences at sites of HIV-1 integration: implications for the mechanism of target DNA binding. Nucleic Acids Res. 42, 5164–5176 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Demeulemeester, J. et al. HIV-1 integrase variants retarget viral integration and are associated with disease progression in a chronic infection cohort. Cell Host Microbe 16, 651–662 (2014).

    CAS  PubMed  Google Scholar 

  58. Luger, K., Mader, A. W., Richmond, R. K., Sargent, D. F. & Richmond, T. J. Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260 (1997).

    CAS  PubMed  Google Scholar 

  59. Pryciak, P. M. & Varmus, H. E. Nucleosomes, DNA-binding proteins, and DNA sequence modulate retroviral integration target site selection. Cell 69, 769–780 (1992).

    CAS  PubMed  Google Scholar 

  60. Pruss, D., Bushman, F. D. & Wolffe, A. P. Human immunodeficiency virus integrase directs integration to sites of severe DNA distortion within the nucleosome core. Proc. Natl Acad. Sci. USA 91, 5913–5917 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, G. P., Ciuffi, A., Leipzig, J., Berry, C. C. & Bushman, F. D. HIV integration site selection: analysis by massively parallel pyrosequencing reveals association with epigenetic modifications. Genome Res. 17, 1186–1194 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Roth, S. L., Malani, N. & Bushman, F. D. Gammaretroviral integration into nucleosomal target DNA in vivo. J. Virol. 85, 7393–7401 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Baller, J. A., Gao, J., Stamenova, R., Curcio, M. J. & Voytas, D. F. A nucleosomal surface defines an integration hotspot for the Saccharomyces cerevisiae Ty1 retrotransposon. Genome Res. 22, 704–713 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Maskell, D. P. et al. Structural basis for retroviral integration into nucleosomes. Nature 523, 366–369 (2015). Together with Wilson et al. (2019), cryo-EM studies elucidate how the PFV intasome engages and locally remodels a nucleosome for integration.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Wilson, M. D. et al. Retroviral integration into nucleosomes through DNA looping and sliding along the histone octamer. Nat. Commun. 10, 4189 (2019).

    PubMed  PubMed Central  Google Scholar 

  66. Benleulmi, M. S. et al. Intasome architecture and chromatin density modulate retroviral integration into nucleosome. Retrovirology 12, 13 (2015).

    PubMed  PubMed Central  Google Scholar 

  67. Serrao, E., Ballandras-Colas, A., Cherepanov, P., Maertens, G. N. & Engelman, A. N. Key determinants of target DNA recognition by retroviral intasomes. Retrovirology 12, 39 (2015).

    PubMed  PubMed Central  Google Scholar 

  68. Engelman, A. N., Maertens, G. N. in Retrovirus-Cell Interactions (ed. Parent, L. J.) Ch. 4, 163–199 (Elsevier Inc., 2018).

  69. Bukrinsky, M. I. et al. Active nuclear import of human immunodeficiency virus type 1 preintegration complexes. Proc. Natl Acad. Sci. USA 89, 6580–6584 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Roe, T., Reynolds, T. C., Yu, G. & Brown, P. O. Integration of murine leukemia virus DNA depends on mitosis. EMBO J. 12, 2099–2108 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Bieniasz, P. D., Weiss, R. A. & McClure, M. O. Cell cycle dependence of foamy retrovirus infection. J. Virol. 69, 7295–7299 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Schroder, A. R. et al. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110, 521–529 (2002). Enabled by the release of the human genome sequence, this study convincingly demonstrates that HIV-1 integration strongly favours active transcription units within gene-dense regions of chromatin.

    CAS  PubMed  Google Scholar 

  73. Wu, X., Li, Y., Crise, B. & Burgess, S. M. Transcription start regions in the human genome are favored targets for MLV integration. Science 300, 1749–1751 (2003). This is the first study to reveal that the gammaretrovirus Moloney murine leukaemia virus favours gene promoter regions for integration.

    CAS  PubMed  Google Scholar 

  74. De Ravin, S. S. et al. Enhancers are major targets for murine leukemia virus vector integration. J. Virol. 88, 4504–4513 (2014).

    PubMed  PubMed Central  Google Scholar 

  75. LaFave, M. C. et al. MLV integration site selection is driven by strong enhancers and active promoters. Nucleic Acids Res. 42, 4257–4269 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Yamashita, M. & Emerman, M. Capsid is a dominant determinant of retrovirus infectivity in nondividing cells. J. Virol. 78, 5670–5678 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. 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 

  78. Matreyek, K. A., Yucel, S. S., Li, X. & Engelman, A. Nucleoporin NUP153 phenylalanine-glycine motifs engage a common binding pocket within the HIV-1 capsid protein to mediate lentiviral infectivity. PLoS Pathog. 9, e1003693 (2013).

    PubMed  PubMed Central  Google Scholar 

  79. Rüegsegger, U., Beyer, K. & Keller, W. Purification and characterization of human cleavage factor Im involved in the 3’ end processing of messenger RNA precursors. J. Biol. Chem. 271, 6107–6113 (1996).

    PubMed  Google Scholar 

  80. Price, A. J. et al. Host cofactors and pharmacologic ligands share an essential interface in HIV-1 capsid that is lost upon disassembly. PLoS Pathog. 10, e1004459 (2014).

    PubMed  PubMed Central  Google Scholar 

  81. Bejarano, D. A. et al. HIV-1 nuclear import in macrophages is regulated by CPSF6-capsid interactions at the nuclear pore complex. eLife 8, e41800 (2019).

    PubMed  PubMed Central  Google Scholar 

  82. Bizhanova, A. & Kaufman, P. D. Close to the edge: heterochromatin at the nucleolar and nuclear peripheries. Biochim. Biophys. Acta Gene Regul. Mech. 1864, 194666 (2021).

    CAS  PubMed  Google Scholar 

  83. Chen, Y. et al. Mapping 3D genome organization relative to nuclear compartments using TSA-Seq as a cytological ruler. J. Cell Biol. 217, 4025–4048 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Francis, A. C. et al. HIV-1 replication complexes accumulate in nuclear speckles and integrate into speckle-associated genomic domains. Nat. Commun. 11, 3505 (2020). This study demonstrates that HIV-1 integration favours regions of chromatin that are located close to nuclear speckles.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Achuthan, V. et al. Capsid-CPSF6 interaction licenses nuclear HIV-1 trafficking to sites of viral DNA integration. Cell Host Microbe 24, 392–404.e398 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Greig, J. A. et al. Arginine-enriched mixed-charge domains provide cohesion for nuclear speckle condensation. Mol. Cell 77, 1237–1250.e1234 (2020).

    CAS  PubMed  Google Scholar 

  87. Li, W. et al. CPSF6-dependent targeting of speckle-associated domains distinguishes primate from nonprimate lentiviral integration. mBio 11, e02254-20 (2020).

    PubMed  PubMed Central  Google Scholar 

  88. Koh, Y. et al. Differential effects of human immunodeficiency virus type 1 capsid and cellular factors nucleoporin 153 and LEDGF/p75 on the efficiency and specificity of viral DNA integration. J. Virol. 87, 648–658 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Ocwieja, K. E. et al. HIV integration targeting: a pathway involving transportin-3 and the nuclear pore protein RanBP2. PLoS Pathog. 7, e1001313 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Müllers, E. The foamy virus Gag proteins: what makes them different? Viruses 5, 1023–1041 (2013).

    PubMed  PubMed Central  Google Scholar 

  91. Tobaly-Tapiero, J. et al. Chromatin tethering of incoming foamy virus by the structural Gag protein. Traffic 9, 1717–1727 (2008).

    CAS  PubMed  Google Scholar 

  92. Lesbats, P. et al. Structural basis for spumavirus GAG tethering to chromatin. Proc. Natl Acad. Sci. USA 114, 5509–5514 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Elis, E., Ehrlich, M., Prizan-Ravid, A., Laham-Karam, N. & Bacharach, E. p12 tethers the murine leukemia virus pre-integration complex to mitotic chromosomes. PLoS Pathog. 8, e1003103 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Schneider, W. M. et al. Viral DNA tethering domains complement replication-defective mutations in the p12 protein of MuLV Gag. Proc. Natl Acad. Sci. USA 110, 9487–9492 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Wanaguru, M., Barry, D. J., Benton, D. J., O’Reilly, N. J. & Bishop, K. N. Murine leukemia virus p12 tethers the capsid-containing pre-integration complex to chromatin by binding directly to host nucleosomes in mitosis. PLoS Pathog. 14, e1007117 (2018).

    PubMed  PubMed Central  Google Scholar 

  96. Busschots, K. et al. The interaction of LEDGF/p75 with integrase is lentivirus-specific and promotes DNA binding. J. Biol. Chem. 280, 17841–17847 (2005).

    CAS  PubMed  Google Scholar 

  97. Cherepanov, P. LEDGF/p75 interacts with divergent lentiviral integrases and modulates their enzymatic activity in vitro. Nucleic Acids Res. 35, 113–124 (2007).

    CAS  PubMed  Google Scholar 

  98. Marshall, H. M. et al. Role of PSIP1/LEDGF/p75 in lentiviral infectivity and integration targeting. PLoS ONE 2, e1340 (2007).

    PubMed  PubMed Central  Google Scholar 

  99. Shun, M. C. et al. LEDGF/p75 functions downstream from preintegration complex formation to effect gene-specific HIV-1 integration. Genes Dev. 21, 1767–1778 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Singh, P. K. et al. LEDGF/p75 interacts with mRNA splicing factors and targets HIV-1 integration to highly spliced genes. Genes Dev. 29, 2287–2297 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Sowd, G. A. et al. A critical role for alternative polyadenylation factor CPSF6 in targeting HIV-1 integration to transcriptionally active chromatin. Proc. Natl Acad. Sci. USA 113, E1054–E1063 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. LeRoy, G. et al. LEDGF and HDGF2 relieve the nucleosome-induced barrier to transcription in differentiated cells. Sci. Adv. 5, eaay3068 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Llano, M. et al. Identification and characterization of the chromatin-binding domains of the HIV-1 integrase interactor LEDGF/p75. J. Mol. Biol. 360, 760–773 (2006).

    CAS  PubMed  Google Scholar 

  104. Turlure, F., Maertens, G., Rahman, S., Cherepanov, P. & Engelman, A. A tripartite DNA-binding element, comprised of the nuclear localization signal and two AT-hook motifs, mediates the association of LEDGF/p75 with chromatin in vivo. Nucleic Acids Res. 34, 1653–1665 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Cherepanov, P., Devroe, E., Silver, P. A. & Engelman, A. Identification of an evolutionarily conserved domain in human lens epithelium-derived growth factor/transcriptional co-activator p75 (LEDGF/p75) that binds HIV-1 integrase. J. Biol. Chem. 279, 48883–48892 (2004).

    CAS  PubMed  Google Scholar 

  106. Maertens, G. et al. LEDGF/p75 is essential for nuclear and chromosomal targeting of HIV-1 integrase in human cells. J. Biol. Chem. 278, 33528–33539 (2003).

    CAS  PubMed  Google Scholar 

  107. Hare, S. et al. A novel co-crystal structure affords the design of gain-of-function lentiviral integrase mutants in the presence of modified PSIP1/LEDGF/p75. PLoS Pathog. 5, e1000259 (2009).

    PubMed  PubMed Central  Google Scholar 

  108. Cherepanov, P. et al. Solution structure of the HIV-1 integrase-binding domain in LEDGF/p75. Nat. Struct. Mol. Biol. 12, 526–532 (2005).

    CAS  PubMed  Google Scholar 

  109. Cherepanov, P., Ambrosio, A. L., Rahman, S., Ellenberger, T. & Engelman, A. Structural basis for the recognition between HIV-1 integrase and transcriptional coactivator p75. Proc. Natl Acad. Sci. USA 102, 17308–17313 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Eidahl, J. O. et al. Structural basis for high-affinity binding of LEDGF PWWP to mononucleosomes. Nucleic Acids Res. 41, 3924–3936 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Wang, H., Farnung, L., Dienemann, C. & Cramer, P. Structure of H3K36-methylated nucleosome-PWWP complex reveals multivalent cross-gyre binding. Nat. Struct. Mol. Biol. 27, 8–13 (2020).

    CAS  PubMed  Google Scholar 

  112. Shun, M. C. et al. Identification and characterization of PWWP domain residues critical for LEDGF/p75 chromatin binding and human immunodeficiency virus type 1 infectivity. J. Virol. 82, 11555–11567 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Tesina, P. et al. Multiple cellular proteins interact with LEDGF/p75 through a conserved unstructured consensus motif. Nat. Commun. 6, 7968 (2015).

    CAS  PubMed  Google Scholar 

  114. De Rijck, J. et al. The BET family of proteins targets moloney murine leukemia virus integration near transcription start sites. Cell Rep. 5, 886–894 (2013).

    PubMed  PubMed Central  Google Scholar 

  115. Gupta, S. S. et al. Bromo- and extraterminal domain chromatin regulators serve as cofactors for murine leukemia virus integration. J. Virol. 87, 12721–12736 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Sharma, A. et al. BET proteins promote efficient murine leukemia virus integration at transcription start sites. Proc. Natl Acad. Sci. USA 110, 12036–12041 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Larue, R. C. et al. Bimodal high-affinity association of Brd4 with murine leukemia virus integrase and mononucleosomes. Nucleic Acids Res. 42, 4868–4881 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. LeRoy, G. et al. Proteogenomic characterization and mapping of nucleosomes decoded by Brd and HP1 proteins. Genome Biol. 13, R68 (2012).

    PubMed  PubMed Central  Google Scholar 

  119. Morinière, J. et al. Cooperative binding of two acetylation marks on a histone tail by a single bromodomain. Nature 461, 664–668 (2009).

    PubMed  Google Scholar 

  120. Rahman, S. et al. The Brd4 extraterminal domain confers transcription activation independent of pTEFb by recruiting multiple proteins, including NSD3. Mol. Cell Biol. 31, 2641–2652 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Aiyer, S. et al. Altering murine leukemia virus integration through disruption of the integrase and BET protein family interaction. Nucleic Acids Res. 42, 5917–5928 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Crowe, B. L. et al. Structure of the Brd4 ET domain bound to a C-terminal motif from γ-retroviral integrases reveals a conserved mechanism of interaction. Proc. Natl Acad. Sci. USA 113, 2086–2091 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Aiyer, S. et al. A common binding motif in the ET domain of BRD3 forms polymorphic structural interfaces with host and viral proteins. Structure https://doi.org/10.1016/j.str.2021.01.010 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  124. Maertens, G. N. B′-protein phosphatase 2A is a functional binding partner of delta-retroviral integrase. Nucleic Acids Res. 44, 364–376 (2016).

    CAS  PubMed  Google Scholar 

  125. Winans, S. et al. The FACT complex promotes avian leukosis virus DNA integration. J. Virol. 91, e00082–17 (2017).

    PubMed  PubMed Central  Google Scholar 

  126. Hertz, E. P. T. et al. A conserved motif provides binding specificity to the PP2A-B56 phosphatase. Mol. Cell 63, 686–695 (2016).

    CAS  PubMed  Google Scholar 

  127. Mitchell, R. S. et al. Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol. 2, E234 (2004).

    PubMed  PubMed Central  Google Scholar 

  128. Gillet, N. A. et al. The host genomic environment of the provirus determines the abundance of HTLV-1-infected T-cell clones. Blood 117, 3113–3122 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Christensen, D. E., Ganser-Pornillos, B. K., Johnson, J. S., Pornillos, O. & Sundquist, W. I. Reconstitution and visualization of HIV-1 capsid-dependent replication and integration in vitro. Science 370, eabc8420 (2020). This study reports reconstitution of HIV-1 reverse transcription and integration in vitro from permeabilized virus particles.

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Bukovsky, A. & Gottlinger, H. Lack of integrase can markedly affect human immunodeficiency virus type 1 particle production in the presence of an active viral protease. J. Virol. 70, 6820–6825 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Engelman, A., Englund, G., Orenstein, J. M., Martin, M. A. & Craigie, R. Multiple effects of mutations in human immunodeficiency virus type 1 integrase on viral replication. J. Virol. 69, 2729–2736 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Elliott, J. L. et al. Integrase-RNA interactions underscore the critical role of integrase in HIV-1 virion morphogenesis. eLife 9, e543111 (2020).

    Google Scholar 

  133. Engelman, A. In vivo analysis of retroviral integrase structure and function. Adv. Virus Res. 52, 411–426 (1999).

    CAS  PubMed  Google Scholar 

  134. Leavitt, A. D., Robles, G., Alesandro, N. & Varmus, H. E. Human immunodeficiency virus type 1 integrase mutants retain in vitro integrase activity yet fail to integrate viral DNA efficiently during infection. J. Virol. 70, 721–728 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Mohammed, K. D., Topper, M. B. & Muesing, M. A. Sequential deletion of the integrase (Gag-Pol) carboxyl terminus reveals distinct phenotypic classes of defective HIV-1. J. Virol. 85, 4654–4666 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Hoyte, A. C. et al. Resistance to pyridine-based inhibitor KF116 reveals an unexpected role of integrase in HIV-1 Gag-Pol polyprotein proteolytic processing. J. Biol. Chem. 292, 19814–19825 (2017).

    CAS  PubMed  Google Scholar 

  137. Dobard, C. W., Briones, M. S. & Chow, S. A. Molecular mechanisms by which human immunodeficiency virus type 1 integrase stimulates the early steps of reverse transcription. J. Virol. 81, 10037–10046 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Tekeste, S. S. et al. Interaction between reverse transcriptase and integrase is required for reverse transcription during HIV-1 replication. J. Virol. 89, 12058–12069 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Fontana, J. et al. Distribution and redistribution of HIV-1 nucleocapsid protein in immature, mature, and integrase-inhibited virions: a role for integrase in maturation. J. Virol. 89, 9765–9780 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Jurado, K. A. et al. Allosteric integrase inhibitor potency is determined through the inhibition of HIV-1 particle maturation. Proc. Natl Acad. Sci. USA 110, 8690–8695 (2013). This is the first study to reveal that ALLINIs potently inhibit HIV-1 maturation.

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Kessl, J. J. et al. HIV-1 integrase binds the viral RNA genome and is essential during virion morphogenesis. Cell 166, 1257–1268 e1212 (2016). This study provides evidence that IN binding to viral RNA has a crucial role in RNA encapsidation, virus particle maturation and HIV-1 infection.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Balakrishnan, M. et al. Non-catalytic site HIV-1 integrase inhibitors disrupt core maturation and induce a reverse transcription block in target cells. PLoS ONE 8, e74163 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Steinrigl, A. et al. Mutations in the catalytic core or the C-terminus of murine leukemia virus (MLV) integrase disrupt virion infectivity and exert diverse effects on reverse transcription. Virology 362, 50–59 (2007).

    CAS  PubMed  Google Scholar 

  144. Wlodawer, A. & Vondrasek, J. Inhibitors of HIV-1 protease: a major success of structure-assisted drug design. Annu. Rev. Biophys. Biomol. Struct. 27, 249–284 (1998).

    CAS  PubMed  Google Scholar 

  145. Pattishall, K. H. in The Search for Antiviral Drugs: Case Histories from Concept to Clinic (eds Adams, J. & Merluzzi, V. J.) 23–43 (Birkhäuser, 1993).

  146. Murray, J. M. et al. Antiretroviral therapy with the integrase inhibitor raltegravir alters decay kinetics of HIV, significantly reducing the second phase. AIDS 21, 2315–2321 (2007).

    CAS  PubMed  Google Scholar 

  147. Sato, M. et al. Novel HIV-1 integrase inhibitors derived from quinolone antibiotics. J. Med. Chem. 49, 1506–1508 (2006).

    CAS  PubMed  Google Scholar 

  148. DeJesus, E. et al. Antiviral activity, pharmacokinetics, and dose response of the HIV-1 integrase inhibitor GS-9137 (JTK-303) in treatment-naive and treatment-experienced patients. J. Acquir. Immune Defic. Syndr. 43, 1–5 (2006).

    CAS  PubMed  Google Scholar 

  149. Tsiang, M. et al. Antiviral activity of bictegravir (GS-9883), a novel potent HIV-1 integrase strand transfer inhibitor with an improved resistance profile. Antimicrob. Agents Chemother. 60, 7086–7097 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Markham, A. Cabotegravir plus rilpivirine: first approval. Drugs 80, 915–922 (2020).

    CAS  PubMed  Google Scholar 

  151. Hassounah, S. A. et al. Antiviral activity of bictegravir and cabotegravir against integrase inhibitor-resistant SIVmac239 and HIV-1. Antimicrob. Agents Chemother. 61, e01695–17 (2017).

    PubMed  PubMed Central  Google Scholar 

  152. Smith, S. J., Zhao, X. Z., Burke, T. R. Jr. & Hughes, S. H. Efficacies of cabotegravir and bictegravir against drug-resistant HIV-1 integrase mutants. Retrovirology 15, 37 (2018).

    PubMed  PubMed Central  Google Scholar 

  153. Zhang, W. W. et al. Accumulation of multiple mutations in vivo confers cross-resistance to new and existing integrase inhibitors. J. Infect. Dis. 218, 1773–1776 (2018).

    PubMed  Google Scholar 

  154. WHO. Update of recommendations on first- and second-line antiretroviral regimens. WHO Policy Brief 1–16 (WHO, 2019).

  155. Hazuda, D. J. et al. Inhibitors of strand transfer that prevent integration and inhibit HIV-1 replication in cells. Science 287, 646–650 (2000). This study reports the initial discovery and characterization of INSTIs.

    CAS  PubMed  Google Scholar 

  156. Grobler, J. A. et al. Diketo acid inhibitor mechanism and HIV-1 integrase: implications for metal binding in the active site of phosphotransferase enzymes. Proc. Natl Acad. Sci. USA 99, 6661–6666 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Espeseth, A. S. et al. HIV-1 integrase inhibitors that compete with the target DNA substrate define a unique strand transfer conformation for integrase. Proc. Natl Acad. Sci. USA 97, 11244–11249 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Valkov, E. et al. Functional and structural characterization of the integrase from the prototype foamy virus. Nucleic Acids Res. 37, 243–255 (2009).

    CAS  PubMed  Google Scholar 

  159. Barski, M. S., Minnell, J. J. & Maertens, G. N. Inhibition of HTLV-1 infection by HIV-1 first- and second-generation integrase strand transfer inhibitors. Front. Microbiol. 10, 1877 (2019).

    PubMed  PubMed Central  Google Scholar 

  160. Koh, Y., Matreyek, K. A. & Engelman, A. Differential sensitivities of retroviruses to integrase strand transfer inhibitors. J. Virol. 85, 3677–3682 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Hare, S. et al. Molecular mechanisms of retroviral integrase inhibition and the evolution of viral resistance. Proc. Natl Acad. Sci. USA 107, 20057–20062 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Hare, S. et al. Structural and functional analyses of the second-generation integrase strand transfer inhibitor dolutegravir (S/GSK1349572). Mol. Pharmacol. 80, 565–572 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Langley, D. R. et al. The terminal (catalytic) adenosine of the HIV LTR controls the kinetics of binding and dissociation of HIV integrase strand transfer inhibitors. Biochemistry 47, 13481–13488 (2008).

    CAS  PubMed  Google Scholar 

  164. Hightower, K. E. et al. Dolutegravir (S/GSK1349572) exhibits significantly slower dissociation than raltegravir and elvitegravir from wild-type and integrase inhibitor-resistant HIV-1 integrase-DNA complexes. Antimicrob. Agents Chemother. 55, 4552–4559 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. White, K. L. et al. Long dissociation of bictegravir from HIV-1 integrase-DNA complexes. Antimicrob. Agents Chemother. 65, e02406–e02420 (2021).

    CAS  PubMed Central  Google Scholar 

  166. Grobler, J. A. & Hazuda, D. J. Resistance to HIV integrase strand transfer inhibitors: in vitro findings and clinical consequences. Curr. Opin. Virol. 8, 98–103 (2014).

    CAS  PubMed  Google Scholar 

  167. Santoro, M. M. et al. Susceptibility to HIV-1 integrase strand transfer inhibitors (INSTIs) in highly treatment-experienced patients who failed an INSTI-based regimen. Int. J. Antimicrob. Agents 56, 106027 (2020).

    CAS  PubMed  Google Scholar 

  168. Ndashimye, E. et al. Accumulation of integrase strand transfer inhibitor resistance mutations confers high-level resistance to dolutegravir in non-B subtype HIV-1 strains from patients failing raltegravir in Uganda. J. Antimicrob. Chemother. 75, 3525–3533 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Rhee, S. Y. et al. A systematic review of the genetic mechanisms of dolutegravir resistance. J. Antimicrob. Chemother. 74, 3135–3149 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Engone-Ondo, J. D. et al. High rate of virological failure and HIV drug resistance in semi-rural Gabon and implications for dolutegravir-based regimen efficacy. J. Antimicrob. Chemother. 76, 1051–1056 (2020).

    Google Scholar 

  171. Christ, F. et al. Rational design of small-molecule inhibitors of the LEDGF/p75-integrase interaction and HIV replication. Nat. Chem. Biol. 6, 442–448 (2010).

    CAS  PubMed  Google Scholar 

  172. Fader, L. D. et al. Discovery of BI 224436, a noncatalytic site integrase inhibitor (NCINI) of HIV-1. ACS Med. Chem. Lett. 5, 422–427 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Tsiang, M. et al. New class of HIV-1 integrase (IN) inhibitors with a dual mode of action. J. Biol. Chem. 287, 21189–21203 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Le Rouzic, E. et al. Dual inhibition of HIV-1 replication by integrase-LEDGF allosteric inhibitors is predominant at the post-integration stage. Retrovirology 10, 144 (2013).

    PubMed  PubMed Central  Google Scholar 

  175. Gupta, K. et al. Allosteric inhibition of human immunodeficiency virus integrase: late block during viral replication and abnormal multimerization involving specific protein domains. J. Biol. Chem. 289, 20477–20488 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Desimmie, B. A. et al. LEDGINs inhibit late stage HIV-1 replication by modulating integrase multimerization in the virions. Retrovirology 10, 57 (2013).

    PubMed  PubMed Central  Google Scholar 

  177. Sharma, A. et al. A new class of multimerization selective inhibitors of HIV-1 integrase. PLoS Pathog. 10, e1004171 (2014).

    PubMed  PubMed Central  Google Scholar 

  178. Deng, N. et al. Allosteric HIV-1 integrase inhibitors promote aberrant protein multimerization by directly mediating inter-subunit interactions: structural and thermodynamic modeling studies. Protein Sci. 25, 1911–1917 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Gupta, K. et al. Structural basis for inhibitor-induced aggregation of HIV Integrase. PLoS Biol. 14, e1002584 (2016).

    PubMed  PubMed Central  Google Scholar 

  180. Koneru, P. C. et al. HIV-1 integrase tetramers are the antiviral target of pyridine-based allosteric integrase inhibitors. eLife 8, e46344 (2019).

    PubMed  PubMed Central  Google Scholar 

  181. Gupta, K. et al. Allosteric HIV integrase inhibitors promote formation of inactive branched polymers via homomeric carboxy-terminal domain interactions. Structure 29, 213–225.e5 (2021).

    CAS  PubMed  Google Scholar 

  182. Amadori, C. et al. The HIV-1 integrase-LEDGF allosteric inhibitor MUT-A: resistance profile, impairment of virus maturation and infectivity but without influence on RNA packaging or virus immunoreactivity. Retrovirology 14, 50 (2017).

    PubMed  PubMed Central  Google Scholar 

  183. Bruggemans, A. et al. GS-9822, a preclinical LEDGIN candidate, displays a block-and-lock phenotype in cell culture. Antimicrob. Agents Chemother. 65, e02328–20 (2021).

    CAS  PubMed Central  Google Scholar 

  184. Feng, L. et al. The competitive interplay between allosteric HIV-1 integrase inhibitor BI/D and LEDGF/p75 during the early stage of HIV-1 replication adversely affects inhibitor potency. ACS Chem. Biol. 11, 1313–1321 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Vranckx, L. S. et al. LEDGIN-mediated inhibition of integrase-LEDGF/p75 interaction reduces reactivation of residual latent HIV. EBioMedicine 8, 248–264 (2016).

    PubMed  PubMed Central  Google Scholar 

  186. Bonnard, D. et al. Structure-function analyses unravel distinct effects of allosteric inhibitors of HIV-1 integrase on viral maturation and integration. J. Biol. Chem. 293, 6172–6186 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Peese, K. M. et al. 5,6,7,8-Tetrahydro-1,6-naphthyridine derivatives as potent HIV-1-integrase-allosteric-site inhibitors. J. Med. Chem. 62, 1348–1361 (2019).

    CAS  PubMed  Google Scholar 

  188. Fenwick, C. et al. Preclinical profile of BI 224436, a novel HIV-1 non-catalytic-site integrase inhibitor. Antimicrob. Agents Chemother. 58, 3233–3244 (2014).

    PubMed  PubMed Central  Google Scholar 

  189. Shkriabai, N. et al. A critical role of the C-terminal segment for allosteric inhibitor-induced aberrant multimerization of HIV-1 integrase. J. Biol. Chem. 289, 26430–26440 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Sutton, G. et al. Assembly intermediates of orthoreovirus captured in the cell. Nat. Commun. 11, 4445 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Hoffman, D. P. et al. Correlative three-dimensional super-resolution and block-face electron microscopy of whole vitreously frozen cells. Science 367, 1–12 (2020).

    Google Scholar 

Download references

Acknowledgements

The authors apologize to colleagues whose work they were unable to cite due to space constraints. This work was supported by US National Institutes of Health grants P50 AI150481 (P.C. and A.N.E.) and R01 AI070042 (A.N.E.), Wellcome Trust Investigator Award 107005/Z/15Z (G.N.M.) and the Francis Crick Institute (P.C), which receives its core funding from Cancer Research UK (FC001061), the UK Medical Research Council (FC001061) and the Wellcome Trust (FC001061).

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Goedele N. Maertens, Alan N. Engelman or Peter Cherepanov.

Ethics declarations

Competing interests

A.N.E. consults for ViiV Healthcare Co. The other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Microbiology thanks H. Aihara, S. Goff and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maertens, G.N., Engelman, A.N. & Cherepanov, P. Structure and function of retroviral integrase. Nat Rev Microbiol 20, 20–34 (2022). https://doi.org/10.1038/s41579-021-00586-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-021-00586-9

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