Expression and reactivation of HIV in a chemokine induced model of HIV latency in primary resting CD4+ T cells

Suha Saleh, Fiona Wightman, Saumya Ramanayake, Marina Alexander, Nitasha Kumar, Gabriela Khoury, Cândida Pereira, Damian Purcell, Paul U Cameron, Sharon R Lewin, Suha Saleh, Fiona Wightman, Saumya Ramanayake, Marina Alexander, Nitasha Kumar, Gabriela Khoury, Cândida Pereira, Damian Purcell, Paul U Cameron, Sharon R Lewin

Abstract

Background: We recently described that HIV latent infection can be established in vitro following incubation of resting CD4+ T-cells with chemokines that bind to CCR7. The main aim of this study was to fully define the post-integration blocks to virus replication in this model of CCL19-induced HIV latency.

Results: High levels of integrated HIV DNA but low production of reverse transcriptase (RT) was found in CCL19-treated CD4+ T-cells infected with either wild type (WT) NL4.3 or single round envelope deleted NL4.3 pseudotyped virus (NL4.3- Δenv). Supernatants from CCL19-treated cells infected with either WT NL4.3 or NL4.3- Δenv did not induce luciferase expression in TZM-bl cells, and there was no expression of intracellular p24. Following infection of CCL19-treated CD4+ T-cells with NL4.3 with enhanced green fluorescent protein (EGFP) inserted into the nef open reading frame (NL4.3- Δnef-EGFP), there was no EGFP expression detected. These data are consistent with non-productive latent infection of CCL19-treated infected CD4+ T-cells. Treatment of cells with phytohemagluttinin (PHA)/IL-2 or CCL19, prior to infection with WT NL4.3, resulted in a mean fold change in unspliced (US) RNA at day 4 compared to day 0 of 21.2 and 1.1 respectively (p = 0.01; n = 5), and the mean expression of multiply spliced (MS) RNA was 56,000, and 5,000 copies/million cells respectively (p = 0.01; n = 5). In CCL19-treated infected CD4+ T-cells, MS-RNA was detected in the nucleus and not in the cytoplasm; in contrast to PHA/IL-2 activated infected cells where MS RNA was detected in both. Virus could be recovered from CCL19-treated infected CD4+ T-cells following mitogen stimulation (with PHA and phorbyl myristate acetate (PMA)) as well as TNFα, IL-7, prostratin and vorinostat.

Conclusions: In this model of CCL19-induced HIV latency, we demonstrate HIV integration without spontaneous production of infectious virus, detection of MS RNA in the nucleus only, and the induction of virus production with multiple activating stimuli. These data are consistent with ex vivo findings from latently infected CD4+ T-cells from patients on combination antiretroviral therapy, and therefore provide further support of this model as an excellent in vitro model of HIV latency.

Figures

Figure 1
Figure 1
High levels of HIV integration with minimal virus production in CCL19-treated infected CD4+ T-cells consistent with latent infection. (A). Schematic diagram of the experimental protocol used for isolation of CD4+ T-cells and HIV infection. Resting CD4+ T-cells were cultured for 2 days with PHA/IL-2, CCL19 or without activation (unactivated). Cells were then infected with WT NL4.3, NL4.3Δenv or NL4.3-Δnef/EGFP or mock for 2 hrs and virus was washed off. The infected cells were cultured with media containing IL-2 (10 IU/mL) for 4 days. Infection of cells with WT NL4.3 (open bars) or NL4.3Δenv (grey bars) was quantified by detection of (B) integrated HIV DNA (C) RT activity (CPM/μl) in culture supernatant or (D) luciferase activity of supernatants using the TZM-bl indicator cell line. In all graphs, the mean (column) and individual data (open symbols) from two donors are shown.
Figure 2
Figure 2
No productive HIV infection in CCL19-treated infected cells. Resting CD4+ T cells were cultured for 2 days with PHA/IL-2, CCL19 or without activation (unactivated). Cells were infected with NL4.3 or NL4.3-Δnef/EGFP for 2 hrs. The virus was washed off and the infected cells were cultured with media containing IL-2 (10 IU/mL) for up to 4 days. Productive infection was detected by flow cytometry of (A) intracellular HIV p24 (Alexa-488) following infection with NL4.3 (left panels; two representative donors). The right hand panels show the corresponding integrated copies of HIV per million cells for the same donors (as the mean of two donors (column) and as individual data (open symbols)). (B) Productive infection was detected by flow cytometry of EGFP expression following infection with NL4.3-Δnef/EGFP (one donor). The integrated copies of HIV per million cells are shown for this donor (right hand panel). SSC = side scatter.
Figure 3
Figure 3
High level of nuclear MS RNA in CCL19-treated latently infected CD4+ T-cells. (A) Schematic diagram of the broad classes of HIV mRNA, including multiply spliced (MS) singly spliced (SS) and unspliced (US) HIV RNA (adapted from [56]). Location of primers and probes used for real-time PCR quantification of US, SS and MS RNA are shown. (B) Resting CD4+ T cells were activated with IL-2/PHA, CCL19 or unactivated and infected with NL4.3 or NL4.3Δenv. The fold increase in US RNA at day 4 post infection (relative to day 0; left panels) and the absolute copies per million cells of MS RNA (right panels) is shown for (B) total RNA following infection with WT NL4.3 (n = 5) and (C and D) nuclear (open) and cytoplasmic (grey) fractions following infection with (C) NL4.3 and (D) NL4.3Δenv. The mean (column) and individual data (open symbols) are shown. The ratio of MS RNA to integrated DNA for each donor is also shown (C and D, far right panel). The detection limit for US RNA and MS RNA was 200 copies/106 cells and is shown as dashed line for MS RNA. * = P < 0.05.
Figure 4
Figure 4
Virus production from latently infected CCL19 stimulated cells. (A) Schematic diagram of the experimental protocol used for infection and restimulation of CCL19-treated resting CD4+ T-cells. Resting CD4+ T-cells were cultured for 2 days with CCL19 and infected with NL4.8 and then restimulated with different activation agents at day 4 post- infection (PI). PHA-stimulated activated PBMCs were added at a ratio of 2:1. The cultures were maintained in IL-2 alone. Supernatant was harvested at day 7 and 10 PI to detect RT activity. (B) RT activity (CPM/μl) was measured following incubation of CCL19-treated infected CD4+ T-cells with (i) PHA/PMA (maximal stimulation) or with DMSO, (ii) TNFα, IL-7, prostratin and the combination of IL-7 with prostratin (expressed as percentage of maximal stimulation), (iii) PHA/PMA or DMSO (from different donors for vorinostat (SAHA) experiments) or (iv) vorionostat (SAHA) expressed as percentage of maximal stimulation (from (iii)). The mean (column) and individual data (open symbols) from four donors are shown. (C) The latently infected cell line ACH2 was also incubated with the same stimuli as in (B) and supernatant collected at day 1, 2 and 3 post stimulation. The mean (column) and individual data (open symbols) from three separate experiments, following activation with vorinostat (SAHA) are shown.

References

    1. Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, Hermankova M, Chadwick K, Margolick J, Quinn TC. et al.Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997;387:183–188. doi: 10.1038/387183a0.
    1. Chun TW, Finzi D, Margolick J, Chadwick K, Schwartz D, Siliciano RF. In vivo fate of HIV-1-infected T cells: quantitative analysis of the transition to stable latency. Nat Med. 1995;1:1284–1290. doi: 10.1038/nm1295-1284.
    1. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R. et al.Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science. 1997;278:1295–1300. doi: 10.1126/science.278.5341.1295.
    1. Lewin SR, Rouzioux C. HIV cure and eradication: how will we get from the laboratory to effective clinical trials? Aids. 2011;25:885–897. doi: 10.1097/QAD.0b013e3283467041.
    1. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel MR, Ghattas G, Brenchley JM. et al.HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15:893–900. doi: 10.1038/nm.1972.
    1. Bosque A, Planelles V. Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells. Blood. 2009;113:58–65. doi: 10.1182/blood-2008-07-168393.
    1. Brooks DG, Hamer DH, Arlen PA, Gao L, Bristol G, Kitchen CM, Berger EA, Zack JA. Molecular characterization, reactivation, and depletion of latent HIV. Immunity. 2003;19:413–423. doi: 10.1016/S1074-7613(03)00236-X.
    1. Plesa G, Dai J, Baytop C, Riley JL, June CH, O'Doherty U. Addition of deoxynucleosides enhances human immunodeficiency virus type 1 integration and 2LTR formation in resting CD4+ T cells. J Virol. 2007;81:13938–13942. doi: 10.1128/JVI.01745-07.
    1. Sahu GK, Lee K, Ji J, Braciale V, Baron S, Cloyd MW. A novel in vitro system to generate and study latently HIV-infected long-lived normal CD4+ T-lymphocytes. Virology. 2006;355:127–137. doi: 10.1016/j.virol.2006.07.020.
    1. Pace MJ, Agosto L, Graf EH, O'Doherty U. HIV reservoirs and latency models. Virology. 2011;411:344–354. doi: 10.1016/j.virol.2010.12.041.
    1. Cameron PU, Saleh S, Sallmann G, Solomon A, Wightman F, Evans VA, Boucher G, Haddad EK, Sekaly RP, Harman AN. et al.Establishment of HIV-1 latency in resting CD4+ T cells depends on chemokine-induced changes in the actin cytoskeleton. Proc Natl Acad Sci USA. 2010;107:16934–16939. doi: 10.1073/pnas.1002894107.
    1. Saleh S, Solomon A, Wightman F, Xhilaga M, Cameron PU, Lewin SR. CCR7 ligands CCL19 and CCL21 increase permissiveness of resting memory CD4+ T cells to HIV-1 infection: a novel model of HIV-1 latency. Blood. 2007;110:4161–4164. doi: 10.1182/blood-2007-06-097907.
    1. Cyster JG. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol. 2005;23:127–159. doi: 10.1146/annurev.immunol.23.021704.115628.
    1. Xie JH, Nomura N, Lu M, Chen SL, Koch GE, Weng Y, Rosa R, Di Salvo J, Mudgett J, Peterson LB. et al.Antibody-mediated blockade of the CXCR3 chemokine receptor results in diminished recruitment of T helper 1 cells into sites of inflammation. J Leukoc Biol. 2003;73:771–780. doi: 10.1189/jlb.1102573.
    1. Yamazaki T, Yang XO, Chung Y, Fukunaga A, Nurieva R, Pappu B, Martin-Orozco N, Kang HS, Ma L, Panopoulos AD. et al.CCR6 regulates the migration of inflammatory and regulatory T cells. J Immunol. 2008;181:8391–8401.
    1. Forster R, Davalos-Misslitz AC, Rot A. CCR7 and its ligands: balancing immunity and tolerance. Nat Rev Immunol. 2008;8:362–371. doi: 10.1038/nri2297.
    1. Chun TW, Nickle DC, Justement JS, Meyers JH, Roby G, Hallahan CW, Kottilil S, Moir S, Mican JM, Mullins JI. et al.Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis. 2008;197:714–720. doi: 10.1086/527324.
    1. Yukl SA, Gianella S, Sinclair E, Epling L, Li Q, Duan L, Choi AL, Girling V, Ho T, Li P. et al.Differences in HIV burden and immune activation within the gut of HIV-positive patients receiving suppressive antiretroviral therapy. J Infect Dis. 2010;202:1553–1561. doi: 10.1086/656722.
    1. North TW, Higgins J, Deere JD, Hayes TL, Villalobos A, Adamson L, Shacklett BL, Schinazi RF, Luciw PA. Viral sanctuaries during highly active antiretroviral therapy in a nonhuman primate model for AIDS. J Virol. 2010;84:2913–2922. doi: 10.1128/JVI.02356-09.
    1. Chun TW, Justement JS, Lempicki RA, Yang J, Dennis G Jr, Hallahan CW, Sanford C, Pandya P, Liu S, McLaughlin M. et al.Gene expression and viral prodution in latently infected, resting CD4+ T cells in viremic versus aviremic HIV-infected individuals. Proc Natl Acad Sci USA. 2003;100:1908–1913. doi: 10.1073/pnas.0437640100.
    1. Han Y, Wind-Rotolo M, Yang HC, Siliciano JD, Siliciano RF. Experimental approaches to the study of HIV-1 latency. Nat Rev Microbiol. 2007;5:95–106. doi: 10.1038/nrmicro1580.
    1. Lassen KG, Ramyar KX, Bailey JR, Zhou Y, Siliciano RF. Nuclear retention of multiply spliced HIV-1 RNA in resting CD4+ T cells. PLoS Pathog. 2006;2:e68. doi: 10.1371/journal.ppat.0020068.
    1. Wang FX, Xu Y, Sullivan J, Souder E, Argyris EG, Acheampong EA, Fisher J, Sierra M, Thomson MM, Najera R. et -7 is a potent and proviral strain-specific inducer of latent HIV-1 cellular reservoirs of infected individuals on virally suppressive HAART. J Clin Invest. 2005;115:128–137.
    1. Kulkosky J, Culnan DM, Roman J, Dornadula G, Schnell M, Boyd MR, Pomerantz RJ. Prostratin: activation of latent HIV-1 expression suggests a potential inductive adjuvant therapy for HAART. Blood. 2001;98:3006–3015. doi: 10.1182/blood.V98.10.3006.
    1. Reuse S, Calao M, Kabeya K, Guiguen A, Gatot JS, Quivy V, Vanhulle C, Lamine A, Vaira D, Demonte D. et al.Synergistic activation of HIV-1 expression by deacetylase inhibitors and prostratin: implications for treatment of latent infection. PLoS One. 2009;4:e6093. doi: 10.1371/journal.pone.0006093.
    1. Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, Margolis DM. Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Hum Retroviruses. 2009;25:207–212. doi: 10.1089/aid.2008.0191.
    1. Contreras X, Schweneker M, Chen CS, McCune JM, Deeks SG, Martin J, Peterlin BM. Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J Biol Chem. 2009;284:6782–6789.
    1. Blazkova J, Trejbalova K, Gondois-Rey F, Halfon P, Philibert P, Guiguen A, Verdin E, Olive D, Van Lint C, Hejnar J, Hirsch I. CpG methylation controls reactivation of HIV from latency. PLoS Pathog. 2009;5:e1000554. doi: 10.1371/journal.ppat.1000554.
    1. Kauder SE, Bosque A, Lindqvist A, Planelles V, Verdin E. Epigenetic regulation of HIV-1 latency by cytosine methylation. PLoS Pathog. 2009;5:e1000495. doi: 10.1371/journal.ppat.1000495.
    1. Laspia MF, Rice AP, Mathews MB. HIV-1 Tat protein increases transcriptional initiation and stabilizes elongation. Cell. 1989;59:283–292. doi: 10.1016/0092-8674(89)90290-0.
    1. Sodroski J, Patarca R, Rosen C, Wong-Staal F, Haseltine W. Location of the trans-activating region on the genome of human T-cell lymphotropic virus type III. Science. 1985;229:74–77. doi: 10.1126/science.2990041.
    1. Bisgrove D, Lewinski M, Bushman F, Verdin E. Molecular mechanisms of HIV-1 proviral latency. Expert Rev Anti Infect Ther. 2005;3:805–814. doi: 10.1586/14787210.3.5.805.
    1. Duverger A, Jones J, May J, Bibollet-Ruche F, Wagner FA, Cron RQ, Kutsch O. Determinants of the establishment of human immunodeficiency virus type 1 latency. J Virol. 2009;83:3078–3093. doi: 10.1128/JVI.02058-08.
    1. Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science. 2009;323:1304–1307. doi: 10.1126/science.1165706.
    1. Audige A, Schlaepfer E, Joller H, Speck RF. Uncoupled anti-HIV and immune-enhancing effects when combining IFN-alpha and IL-7. J Immunol. 2005;175:3724–3736.
    1. Ducrey-Rundquist O, Guyader M, Trono D. Modalities of interleukin-7-induced human immunodeficiency virus permissiveness in quiescent T lymphocytes. J Virol. 2002;76:9103–9111. doi: 10.1128/JVI.76.18.9103-9111.2002.
    1. Alcami J, Lain de, Lera T, Folgueira L, Pedraza MA, Jacque JM, Bachelerie F, Noriega AR, Hay RT, Harrich D, Gaynor RB. et al.Absolute dependence on kappa B responsive elements for initiation and Tat-mediated amplification of HIV transcription in blood CD4 T lymphocytes. Embo J. 1995;14:1552–1560.
    1. Coiras M, Lopez-Huertas MR, Rullas J, Mittelbrunn M, Alcami J. Basal shuttle of NF-kappaB/I kappaB alpha in resting T lymphocytes regulates HIV-1 LTR dependent expression. Retrovirology. 2007;4:56. doi: 10.1186/1742-4690-4-56.
    1. Hiscott J, Kwon H, Genin P. Hostile takeovers: viral appropriation of the NF-kappaB pathway. J Clin Invest. 2001;107:143–151. doi: 10.1172/JCI11918.
    1. Selliah N, Zhang M, DeSimone D, Kim H, Brunner M, Ittenbach RF, Rui H, Cron RQ, Finkel TH. The gammac-cytokine regulated transcription factor, STAT5, increases HIV-1 production in primary CD4 T cells. Virology. 2006;344:283–291. doi: 10.1016/j.virol.2005.09.063.
    1. Crotti A, Lusic M, Lupo R, Lievens PM, Liboi E, Della Chiara G, Tinelli M, Lazzarin A, Patterson BK, Giacca M. et al.Naturally occurring C-terminally truncated STAT5 is a negative regulator of HIV-1 expression. Blood. 2007;109:5380–5389. doi: 10.1182/blood-2006-08-042556.
    1. Alfano M, Poli G. Role of cytokines and chemokines in the regulation of innate immunity and HIV infection. Mol Immunol. 2005;42:161–182. doi: 10.1016/j.molimm.2004.06.016.
    1. Yang HC, Xing S, Shan L, O'Connell K, Dinoso J, Shen A, Zhou Y, Shrum CK, Han Y, Liu JO. et al.Small-molecule screening using a human primary cell model of HIV latency identifies compounds that reverse latency without cellular activation. J Clin Invest. 2009;119:3473–3486.
    1. Osborn L, Kunkel S, Nabel GJ. Tumor necrosis factor alpha and interleukin 1 stimulate the human immunodeficiency virus enhancer by activation of the nuclear factor kappa B. Proc Natl Acad Sci USA. 1989;86:2336–2340. doi: 10.1073/pnas.86.7.2336.
    1. Folks TM, Clouse KA, Justement J, Rabson A, Duh E, Kehrl JH, Fauci AS. Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci USA. 1989;86:2365–2368. doi: 10.1073/pnas.86.7.2365.
    1. Burnett JC, Lim KI, Calafi A, Rossi JJ, Schaffer DV, Arkin AP. Combinatorial latency reactivation for HIV-1 subtypes and variants. J Virol. pp. 5958–5974.
    1. Helseth E, Kowalski M, Gabuzda D, Olshevsky U, Haseltine W, Sodroski J. Rapid complementation assays measuring replicative potential of human immunodeficiency virus type 1 envelope glycoprotein mutants. J Virol. 1990;64:2416–2420.
    1. Goff S, Traktman P, Baltimore D. Isolation and properties of Moloney murine leukemia virus mutants: use of a rapid assay for release of virion reverse transcriptase. J Virol. 1981;38:239–248.
    1. Lewin SR, Murray JM, Solomon A, Wightman F, Cameron PU, Purcell DJ, Zaunders JJ, Grey P, Bloch M, Smith D. et al.Virologic determinants of success after structured treatment interruptions of antiretrovirals in acute HIV-1 infection. J Acquir Immune Defic Syndr. 2008;47:140–147. doi: 10.1097/QAI.0b013e31815dbf7f.
    1. O'Doherty U, Swiggard WJ, Jeyakumar D, McGain D, Malim MH. A sensitive, quantitative assay for human immunodeficiency virus type 1 integration. J Virol. 2002;76:10942–10950. doi: 10.1128/JVI.76.21.10942-10950.2002.
    1. Butler SL, Hansen MS, Bushman FD. A quantitative assay for HIV DNA integration in vivo. Nat Med. 2001;7:631–634. doi: 10.1038/87979.
    1. Zhang Z, Schuler T, Zupancic M, Wietgrefe S, Staskus KA, Reimann KA, Reinhart TA, Rogan M, Cavert W, Miller CJ. et al.Sexual transmission and propagation of SIV and HIV in resting and activated CD4+ T cells. Science. 1999;286:1353–1357. doi: 10.1126/science.286.5443.1353.
    1. Lewin SR, Vesanen M, Kostrikis L, Hurley A, Duran M, Zhang L, Ho DD, Markowitz M. Use of real-time PCR and molecular beacons to detect virus replication in human immunodeficiency virus type 1-infected individuals on prolonged effective antiretroviral therapy. J Virol. 1999;73:6099–6103.
    1. Pereira CF, Ellenberg PC, Jones KL, Fernandez TL, Smyth RP, Hawkes DJ, Hijnen M, Vivet-Boudou V, Marquet R, Johnson I, Mak J. Labeling of multiple HIV-1 proteins with the biarsenical-tetracysteine system. PLoS One. 2011;6:e17016. doi: 10.1371/journal.pone.0017016.
    1. Yang JY, Schwartz A, Henderson EE. Inhibition of HIV-1 latency reactivation by dehydroepiandrosterone (DHEA) and an analog of DHEA. AIDS Res Hum Retroviruses. 1993;9:747–754. doi: 10.1089/aid.1993.9.747.
    1. Purcell DF, Martin MA. Alternative splicing of human immunodeficiency virus type 1 mRNA modulates viral protein expression, replication, and infectivity. J Virol. 1993;67:6365–6378.

Source: PubMed

3
Abonner