1887

Abstract

CRISPR/dCas9-based activation systems (CRISPRa) enable sequence-specific gene activation and are therefore of particular interest for the ‘shock and kill’ cure approach against HIV-1 infections. This approach aims to activate the latent HIV-1 proviruses in infected cells and subsequently kill these cells. Several CRISPRa systems have been shown to specifically and effectively activate latent HIV-1 when targeted to the HIV-1 5′LTR promoter, making them a promising ‘shock’ strategy. Here, we aimed to evaluate the dCas9-VPR system for its applicability in reversing HIV-1 latency and identify the optimal gRNA target site in the HIV-1 5′LTR promoter leading to the strongest activation of the provirus with this system. We systematically screened the HIV-1 promoter by selecting 14 specific gRNAs that cover almost half of the HIV-1 promoter from the 3′ half of the U3 until the beginning of the R region. Screening in several latently HIV-1 infected cell lines showed that dCas9-VPR leads to a high activation of HIV-1 and that gRNA-V and -VII induce the strongest activation of replication competent latent provirus. This data indicates that the optimal activation region in the HIV-1 promoter for the dCas9-VPR system is located −165 to −106 bp from the transcription start site and that it is consistent with the optimal activation region reported for other CRISPRa systems. Our data demonstrates that the dCas9-VPR system is a powerful tool for HIV-1 activation and could be harnessed for the ‘shock and kill’ cure approach.

Funding
This study was supported by the:
  • Gilead Sciences (Award 00408)
    • Principle Award Recipient: KarinJ. Metzner
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001754
2022-06-07
2024-04-19
Loading full text...

Full text loading...

References

  1. WHO Data and statistics; 2020 https://www.who.int/gho/hiv/en/
  2. Chun TW, Finzi D, Margolick J, Chadwick K, Schwartz D et al. In vivo fate of HIV-1-infected T cells: quantitative analysis of the transition to stable latency. Nat Med 1995; 1:1284–1290 [View Article] [PubMed]
    [Google Scholar]
  3. Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 1997; 387:183–188 [View Article] [PubMed]
    [Google Scholar]
  4. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 1997; 278:1295–1300 [View Article] [PubMed]
    [Google Scholar]
  5. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 1999; 5:512–517 [View Article] [PubMed]
    [Google Scholar]
  6. Chun TW, Davey RT Jr, Ostrowski M, Shawn Justement J, Engel D et al. Relationship between pre-existing viral reservoirs and the re-emergence of plasma viremia after discontinuation of highly active anti-retroviral therapy. Nat Med 2000; 6:757–761 [View Article] [PubMed]
    [Google Scholar]
  7. Wong JK, Hezareh M, Günthard HF, Havlir DV, Ignacio CC et al. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997; 278:1291–1295 [View Article] [PubMed]
    [Google Scholar]
  8. Bachmann N, von Siebenthal C, Vongrad V, Turk T, Neumann K et al. Determinants of HIV-1 reservoir size and long-term dynamics during suppressive ART. Nat Commun 2019; 10:3193 [View Article] [PubMed]
    [Google Scholar]
  9. Archin NM, Margolis DM. Emerging strategies to deplete the HIV reservoir. Curr Opin Infect Dis 2014; 27:29–35 [View Article] [PubMed]
    [Google Scholar]
  10. Rasmussen TA, Lewin SR. Shocking HIV out of hiding: where are we with clinical trials of latency reversing agents?. Curr Opin HIV AIDS 2016; 11:394–401 [View Article] [PubMed]
    [Google Scholar]
  11. Razooky BS, Pai A, Aull K, Rouzine IM, Weinberger LS. A hardwired HIV latency program. Cell 2015; 160:990–1001 [View Article] [PubMed]
    [Google Scholar]
  12. Xiao Q, Guo D, Chen S. Application of CRISPR/Cas9-based gene editing in HIV-1/AIDS therapy. Front Cell Infect Microbiol 2019; 9:69 [View Article] [PubMed]
    [Google Scholar]
  13. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 2013; 154:442–451 [View Article] [PubMed]
    [Google Scholar]
  14. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013; 152:1173–1183 [View Article] [PubMed]
    [Google Scholar]
  15. Darcis G, Das AT, Berkhout B. Tackling HIV persistence: pharmacological versus CRISPR-based shock strategies. Viruses 2018; 10:E157 [View Article] [PubMed]
    [Google Scholar]
  16. La Russa MF, Qi LS. The new state of the art: Cas9 for gene activation and repression. Mol Cell Biol 2015; 35:3800–3809 [View Article] [PubMed]
    [Google Scholar]
  17. Chavez A, Tuttle M, Pruitt BW, Ewen-Campen B, Chari R et al. Comparison of Cas9 activators in multiple species. Nat Methods 2016; 13:563–567 [View Article] [PubMed]
    [Google Scholar]
  18. Zhang Y, Yin C, Zhang T, Li F, Yang W et al. CRISPR/gRNA-directed synergistic activation mediator (SAM) induces specific, persistent and robust reactivation of the HIV-1 latent reservoirs. Sci Rep 2015; 5:16277 [View Article] [PubMed]
    [Google Scholar]
  19. Saayman SM, Lazar DC, Scott TA, Hart JR, Takahashi M et al. Potent and targeted activation of latent HIV-1 using the CRISPR/dCas9 activator complex. Mol Ther 2016; 24:488–498 [View Article] [PubMed]
    [Google Scholar]
  20. Limsirichai P, Gaj T, Schaffer DV. CRISPR-mediated activation of latent HIV-1 expression. Mol Ther 2016; 24:499–507 [View Article] [PubMed]
    [Google Scholar]
  21. Ji H, Jiang Z, Lu P, Ma L, Li C et al. Specific reactivation of latent HIV-1 by dCas9-suntag-VP64-mediated Guide RNA targeting the HIV-1 promoter. Mol Ther 2016; 24:508–521 [View Article] [PubMed]
    [Google Scholar]
  22. Bialek JK, Dunay GA, Voges M, Schäfer C, Spohn M et al. Targeted HIV-1 latency reversal using CRISPR/Cas9-derived transcriptional activator systems. PLoS One 2016; 11:e0158294 [View Article] [PubMed]
    [Google Scholar]
  23. Scott TA, O’Meally D, Grepo NA, Soemardy C, Lazar DC et al. Broadly active zinc finger protein-guided transcriptional activation of HIV-1. Mol Ther Methods Clin Dev 2021; 20:18–29 [View Article] [PubMed]
    [Google Scholar]
  24. Chavez A, Scheiman J, Vora S, Pruitt BW, Tuttle M et al. Highly efficient Cas9-mediated transcriptional programming. Nat Methods 2015; 12:326–328 [View Article] [PubMed]
    [Google Scholar]
  25. Perez-Pinera P, Kocak DD, Vockley CM, Adler AF, Kabadi AM et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat Methods 2013; 10:973–976 [View Article] [PubMed]
    [Google Scholar]
  26. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 2015; 517:583–588 [View Article] [PubMed]
    [Google Scholar]
  27. Platt EJ, Wehrly K, Kuhmann SE, Chesebro B, Kabat D. Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1. J Virol 1998; 72:2855–2864 [View Article] [PubMed]
    [Google Scholar]
  28. Derdeyn CA, Decker JM, Sfakianos JN, Wu X, O’Brien WA et al. Sensitivity of human immunodeficiency virus type 1 to the fusion inhibitor T-20 is modulated by coreceptor specificity defined by the V3 loop of gp120. J Virol 2000; 74:8358–8367 [View Article] [PubMed]
    [Google Scholar]
  29. Wei X, Decker JM, Liu H, Zhang Z, Arani RB et al. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 2002; 46:1896–1905 [View Article] [PubMed]
    [Google Scholar]
  30. Takeuchi Y, McClure MO, Pizzato M. Identification of gammaretroviruses constitutively released from cell lines used for human immunodeficiency virus research. J Virol 2008; 82:12585–12588 [View Article] [PubMed]
    [Google Scholar]
  31. Platt EJ, Bilska M, Kozak SL, Kabat D, Montefiori DC. Evidence that ecotropic murine leukemia virus contamination in TZM-bl cells does not affect the outcome of neutralizing antibody assays with human immunodeficiency virus type 1. J Virol 2009; 83:8289–8292 [View Article] [PubMed]
    [Google Scholar]
  32. Jordan A, Bisgrove D, Verdin E. HIV reproducibly establishes a latent infection after acute infection of T cells in vitro. EMBO J 2003; 22:1868–1877 [View Article] [PubMed]
    [Google Scholar]
  33. Clouse KA, Powell D, Washington I, Poli G, Strebel K et al. Monokine regulation of human immunodeficiency virus-1 expression in a chronically infected human T cell clone. Journal of Immunology 1950; 142:431–438
    [Google Scholar]
  34. Folks TM, Clouse KA, Justement J, Rabson A, Duh E et al. Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci U S A 1989; 86:2365–2368 [View Article] [PubMed]
    [Google Scholar]
  35. Russell JN, Clements JE, Gama L. Quantitation of gene expression in formaldehyde-fixed and fluorescence-activated sorted cells. PLoS One 2013; 8:e73849 [View Article] [PubMed]
    [Google Scholar]
  36. Strouvelle VP, Braun DL, Vongrad V, Scherrer AU, Kok YL et al. No effect of pegylated interferon-α on total HIV-1 DNA load in HIV-1/HCV coinfected patients. J Infect Dis 2018; 217:1883–1888 [View Article] [PubMed]
    [Google Scholar]
  37. Roscic-Mrkic B, Fischer M, Leemann C, Manrique A, Gordon CJ et al. RANTES (CCL5) uses the proteoglycan CD44 as an auxiliary receptor to mediate cellular activation signals and HIV-1 enhancement. Blood 2003; 102:1169–1177 [View Article] [PubMed]
    [Google Scholar]
  38. Fischer M, Joos B, Hirschel B, Bleiber G, Weber R et al. Cellular viral rebound after cessation of potent antiretroviral therapy predicted by levels of multiply spliced HIV-1 RNA encoding nef. J Infect Dis 2004; 190:1979–1988 [View Article] [PubMed]
    [Google Scholar]
  39. Fernandez G, Zeichner SL. Cell line-dependent variability in HIV activation employing DNMT inhibitors. Virol J 2010; 7:266 [View Article] [PubMed]
    [Google Scholar]
  40. Tanenbaum ME, Gilbert LA, Qi LS, Weissman JS, Vale RD. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 2014; 159:635–646 [View Article] [PubMed]
    [Google Scholar]
  41. Ehrke-Schulz E, Schiwon M, Leitner T, Dávid S, Bergmann T et al. CRISPR/Cas9 delivery with one single adenoviral vector devoid of all viral genes. Sci Rep 2017; 7:17113 [View Article] [PubMed]
    [Google Scholar]
  42. Dong W, Kantor B. Lentiviral vectors for delivery of gene-editing systems based on CRISPR/Cas: current state and perspectives. Viruses 2021; 13:1288 [View Article] [PubMed]
    [Google Scholar]
  43. Dampier W, Nonnemacher MR, Sullivan NT, Jacobson JM, Wigdahl B. HIV excision utilizing CRISPR/Cas9 technology: attacking the proviral quasispecies in reservoirs to achieve a cure. MOJ Immunol 2014; 1:00022 [View Article] [PubMed]
    [Google Scholar]
  44. Roychoudhury P, De Silva Feelixge H, Reeves D, Mayer BT, Stone D et al. Viral diversity is an obligate consideration in CRISPR/Cas9 designs for targeting the HIV reservoir. BMC Biol 2018; 16:75 [View Article] [PubMed]
    [Google Scholar]
  45. Zhang XH, Tee LY, Wang XG, Huang QS, Yang SH. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids 2015; 4:e264 [View Article] [PubMed]
    [Google Scholar]
  46. Link RW, Nonnemacher MR, Wigdahl B, Dampier W. Prediction of human immunodeficiency virus type 1 subtype-specific off-target effects arising from CRISPR-Cas9 gene editing therapy. CRISPR J 2018; 1:294–302 [View Article] [PubMed]
    [Google Scholar]
  47. Zhang Y, Arango G, Li F, Xiao X, Putatunda R et al. Comprehensive off-target analysis of dCas9-SAM-mediated HIV reactivation via long noncoding RNA and mRNA profiling. BMC Med Genomics 2018; 11:78 [View Article] [PubMed]
    [Google Scholar]
  48. Symons J, Chopra A, Malatinkova E, De Spiegelaere W, Leary S et al. HIV integration sites in latently infected cell lines: evidence of ongoing replication. Retrovirology 2017; 14:2 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001754
Loading
/content/journal/jgv/10.1099/jgv.0.001754
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error