The Tat Inhibitor Didehydro-Cortistatin A Prevents HIV-1 Reactivation from Latency

Guillaume Mousseau, Cari F Kessing, Rémi Fromentin, Lydie Trautmann, Nicolas Chomont, Susana T Valente, Guillaume Mousseau, Cari F Kessing, Rémi Fromentin, Lydie Trautmann, Nicolas Chomont, Susana T Valente

Abstract

Antiretroviral therapy (ART) inhibits HIV-1 replication, but the virus persists in latently infected resting memory CD4(+) T cells susceptible to viral reactivation. The virus-encoded early gene product Tat activates transcription of the viral genome and promotes exponential viral production. Here we show that the Tat inhibitor didehydro-cortistatin A (dCA), unlike other antiretrovirals, reduces residual levels of viral transcription in several models of HIV latency, breaks the Tat-mediated transcriptional feedback loop, and establishes a nearly permanent state of latency, which greatly diminishes the capacity for virus reactivation. Importantly, treatment with dCA induces inactivation of viral transcription even after its removal, suggesting that the HIV promoter is epigenetically repressed. Critically, dCA inhibits viral reactivation upon CD3/CD28 or prostratin stimulation of latently infected CD4(+) T cells from HIV-infected subjects receiving suppressive ART. Our results suggest that inclusion of a Tat inhibitor in current ART regimens may contribute to a functional HIV-1 cure by reducing low-level viremia and preventing viral reactivation from latent reservoirs.

Importance: Antiretroviral therapy (ART) reduces HIV-1 replication to very low levels, but the virus persists in latently infected memory CD4(+) T cells, representing a long-lasting source of resurgent virus upon ART interruption. Based on the mode of action of didehydro-cortistatin A (dCA), a Tat-dependent transcription inhibitor, our work highlights an alternative approach to current HIV-1 eradication strategies to decrease the latent reservoir. In our model, dCA blocks the Tat feedback loop initiated after low-level basal reactivation, blocking transcriptional elongation and hence viral production from latently infected cells. Therefore, dCA combined with ART would be aimed at delaying or halting ongoing viral replication, reactivation, and replenishment of the latent viral reservoir. Thus, the latent pool of cells in an infected individual would be stabilized, and death of the long-lived infected memory T cells would result in a continuous decay of this pool over time, possibly culminating in the long-awaited sterilizing cure.

Copyright © 2015 Mousseau et al.

Figures

FIG 1
FIG 1
dCA inhibits HIV reactivation after TCR stimulation from CD4+ T cells isolated from virally suppressed subjects. (A) CD4+ T cells were isolated from PBMCs from nine virally suppressed infected individuals carefully selected that did not display spontaneous viral production upon in vitro culture to better reflect latency. Activation of viral production from latency with anti-CD3/CD28 beads was performed in the presence of ARVs with or without 100 nM dCA. Viral genomic RNAs from viral particles released in the supernatants were extracted 6 days later and analyzed by ultrasensitive RT-qPCR. NS, nonstimulated; ND, not detected. (B) Summary of the nine subjects. The two-tailed paired t test was used for statistical comparisons.
FIG 2
FIG 2
dCA inhibits residual transcription in HIV-1 latently infected cell line models. (A) dCA inhibits NL4-3 virus expression to undetectable levels in HeLa-CD4 cells (left panel). Latently infected HeLa-CD4 cells were treated with DMSO control or dCA for 239 days (100 nM dCA used from 0 to 118 days and then at 10 nM afterward). dCA treatment was stopped at day 24 (TS1) and day 103 (TS2). TS, treatment stop. Capsid p24 in the supernatants was assayed in an ELISA (detection limit, 3.1 pg/ml). Data are representative of two independent experiments (right panel). For analysis of viral mRNA expression, cDNAs from total RNA extracted at day 239 were quantified by RT-qPCR using primers to the Vpr region. Results were normalized as number of viral mRNA copies per GAPDH mRNA. Viral mRNA generated in the DMSO control was set to 100%. Data are representative of two analyses (days 129 and 239). RT-qPCR data are reported as means ± SD. (B) dCA inhibits viral production in the OM-10.1 cell line to almost-undetectable levels (left panel). OM-10.1 cells were split and treated on average every 3 days in the presence of ARVs with or without dCA. Capsid production was quantified via a p24 ELISA. Data are representative of four independent experiments with treatment of cells ranging from 42 to 226 days (right panel). For analysis of viral mRNA expression in dCA-treated OM-10.1 cells, cDNAs from total RNA (day 197) were quantified, normalized, and are reported as described for panel A using primers to the Gag-Pol region. Data are representative of three analyses at days 102, 163, and 197. (C and D) Effects of dCA on J-Lat 6.3 and 10.6 clones (left panels). Cells were split and treated on average every 3 days with or without 10 nM dCA. Capsid production was quantified via a p24 ELISA, and results for the DMSO controls were set to 100%. Data are reported as means ± SD of two or three independent experiments, respectively. Significant effects of dCA (****, P < 0.0001) and time (****, P < 0.0001) were determined by a two-way repeated-measures ANOVA with Bonferroni correction post hoc (n = 3 per group) (right panels). dCA inhibited viral transcription in J-Lat clones. Viral mRNA levels (day 9) were quantified, normalized, and are reported as described for panel A, using Gag-Pol primers. Data are representative of two independent experiments.
FIG 3
FIG 3
dCA reduces RNAP II recruitment to the HIV promoter and inhibits viral transcription elongation from latently infected HeLa-CD4 cells. (A) Schematic representation of the HIV genome and primer localizations. (B) RNAP II ChIP results with latently infected HeLa-CD4 cells treated long term with DMSO or dCA, at day 172. As controls, DMSO-treated cells were activated with TNF-α for 8 h or inhibited with α-amanitin (α-ama) for 48 h. Data are presented as percentages of input, with the average IgG background subtracted. Data for RT-qPCR (using the indicated primers) are reported as means ± SD. Results are representative of two independent ChIP assays. (C) GAPDH was used as the reference gene, with amplicons to the promoter or ORF.
FIG 4
FIG 4
dCA inhibits viral reactivation from latently infected HeLa-CD4 cells. (A) PMA-iono treatment failed to reactivate virus from dCA-mediated latency in HeLa-CD4 cells. Cells treated with DMSO or dCA at 10 nM were activated or not for 24 h with a combination of PMA and iono. Data are presented as means ± SD of two independent experiments performed at days 131 and 152 post-dCA treatment (n = 2). (B) Exogenous Tat reactivates virus from latency in dCA-treated HeLa-CD4 cells. Cells were evaluated at day 125 post-dCA treatment in the presence or absence of dCA by transfecting an empty vector control or a Tat-Flag-expressing plasmid, and p24 production was determined via an ELISA. Results are representative of three independent transfection experiments.
FIG 5
FIG 5
dCA inhibits viral reactivation from several models of latency. (A) dCA-mediated viral latency in OM-10.1 cells is refractory to reactivation by SAHA, TNF-α, or prostratin. Cells (DMSO controls or treated with dCA at 10 nM) (shown in Fig. 2B) were activated in the presence of ARVs with SAHA or TNF-α for 24 h or prostratin (pros) for 9 h, plus 100 nM dCA for the dCA-treated cells. Supernatant was collected at the end of the activation period and analyzed in a p24 ELISA. Data are presented as means ± SD of two independent activation experiments performed at days 219 and 226 post-dCA treatment (n = 5). (B and C) dCA inhibits viral reactivation from J-Lat 10.6 and 6.3 clones. At day 0, these clones were activated with SAHA, TNF-α, or prostratin in the presence or absence of 100 nM dCA for 24 h. SAHA was not able to activate J-Lat 6.3 above the limit of detection of the assay. Viral production was analyzed and is presented as described for panel in A, as means of two independent activation experiments (n = 4). Percentages represent the percent inhibition. The two-tailed paired t test was used for statistical comparisons. ND, not detectable.
FIG 6
FIG 6
dCA does not affect HIV-1 transcription in the ACH-2 or U1 models of HIV latency. (A and B) ACH-2 and U1 cells were infected with a virus with a mutant TAR or Tat, respectively (left panels). To analyze the effect of dCA on viral production in ACH-2 and U1 cells, cells were split and treated every 3 days on average with or without 10 nM dCA. Capsid production was quantified in a p24 ELISA. Data were normalized for each point to results with the DMSO control (as 100%) and are presented as means ± SD of three independent experiments. For analysis of the effects of dCA and time, a two-way repeated-measures ANOVA was performed. For ACH-2 cells (A), significant effects of dCA treatment (**, P = 0.0037) and time (***, P < 0.0002) are shown. For U1 cells (B), significant effects of dCA treatment (**, P = 0.0065) are shown. After post hoc Bonferroni correction (n = 3 per group), the statistical significance of results is indicated as follows: NS, nonsignificant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 (right panels). dCA does not inhibit viral transcription in ACH-2 or U1 cells. On the indicated day, viral mRNA was extracted and analyzed by RT-qPCR using Gag-Pol primers. RT-qPCR data are presented as means ± SD and are representative of at least two independent experiments. (C and D) dCA does not inhibit viral reactivation in ACH-2 or U1 cells. Cells were grown for 15 days with ARVs and in the presence or absence of dCA at 10 nM. At days 9 and 15, DMSO-treated (blue) or dCA-treated (red) cells and also treated with ARVs were activated with SAHA, TNF-α, or prostratin for 24 h in the presence or absence of 100 nM dCA. Supernatant was collected at the end of the activation period and analyzed in a p24 ELISA. Data are presented as means ± SD of two independent activation experiments performed at days 9 and 15 (n = 4) except for U1 cells (SAHA and prostratin treatments, for which n = 2, due to the use of a different activation protocol at day 9). Percentages indicate the percent inhibition. The two-tailed paired t test was used for statistical comparisons. ND, not detectable.
FIG 7
FIG 7
dCA maintains a state of latency and blocks reactivation in primary expanded CD4+ T cells. (A) Schematic of generation of expanded CD4+ T cells and reactivation. CD4+ T cells were sorted from patient PBMCs and initially expanded with 1 µg/ml PHA, 100 U/ml IL-2, and irradiated autologous feeder PBMCs. Cells were then grown for 22 days in the presence of IL-2 and either ARVs alone or ARVs plus 100 nM dCA. At day 22, cells were split into 6 groups and either stimulated with prostratin or nonstimulated, with treatment stopped (TS) or continued for an additional 6 days before measuring HIV RNA viral production. (B) Limited viral rebound upon dCA removal in expanded primary CD4+ T cells derived from patients A and B. At day 22, the ARVs or ARVs plus dCA cultured CD4+ T cells were washed, and all drugs were removed. Particle-associated HIV genomic RNAs were quantified by RT-qPCR at day 6 after TS (day 28). (C) Inhibition of viral reactivation upon prostratin stimulation in primary CD4+ T cells expanded in the presence of dCA. At day 22, the ARVs or ARVs plus dCA cultured CD4+ T cells were stimulated with prostratin or left unstimulated. Quantification was done as described for panel B (day 28). ND, nondetected. Data are presented as means ± standard errors of the means (n = 2).
FIG 8
FIG 8
Hypothetical approach to a functional HIV cure. (1) Upon HIV infection, there is a sharp increase of the viral load in circulating plasma of infected individuals. (2) The viral load sharply decreases to below the limit of detection (+ T cell reactivation (4). The addition of a Tat inhibitor such as dCA to an ART regimen could promote and maintain a state of latency, possibly allowing for ART interruption without viral rebound. dCA may also prevent reservoir replenishment. With time, patients may potentially observe a reduction in the size of the viral reservoir and relief from chronic inflammation caused by ongoing low-level virus production.

References

    1. Chun TW, Finzi D, Margolick J, Chadwick K, Schwartz D, Siliciano RF. 1995. In vivo fate of HIV-1-infected T cells: quantitative analysis of the transition to stable latency. Nat Med 1:1284–1290. doi:10.1038/nm1295-1284.
    1. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, Lloyd AL, Nowak MA, Fauci AS. 1997. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A 94:13193–13197. doi:10.1073/pnas.94.24.13193.
    1. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF. 1997. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278:1295–1300. doi:10.1126/science.278.5341.1295.
    1. Wong JK, Hezareh M, Günthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD. 1997. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278:1291–1295. doi:10.1126/science.278.5341.1291.
    1. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, Smith K, Lisziewicz J, Lori F, Flexner C, Quinn TC, Chaisson RE, Rosenberg E, Walker B, Gange S, Gallant J, Siliciano RF. 1999. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 5:512–517. doi:10.1038/8394.
    1. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, Kovacs C, Gange SJ, Siliciano RF. 2003. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med 9:727–728. doi:10.1038/nm880.
    1. Buzón MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, Gatell JM, Domingo P, Paredes R, Sharkey M, Palmer S, Stevenson M, Clotet B, Blanco J, Martinez-Picado J. 2010. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 16:460–465. doi:10.1038/nm.2111.
    1. Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG, Beilman GJ, Khoruts A, Thorkelson A, Schmidt TE, Anderson J, Perkey K, Stevenson M, Perelson AS, Douek DC, Haase AT, Schacker TW. 2014. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci U S A 111:2307–2312. doi:10.1073/pnas.1318249111.
    1. Hatano H, Strain MC, Scherzer R, Bacchetti P, Wentworth D, Hoh R, Martin JN, McCune JM, Neaton JD, Tracy RP, Hsue PY, Richman DD, Deeks SG. 2013. Increase in 2-long terminal repeat circles and decrease in d-dimer after raltegravir intensification in patients with treated HIV infection: a randomized, placebo-controlled trial. J Infect Dis 208:1436–1442. doi:10.1093/infdis/jit453.
    1. Besson GJ, McMahon D, Maldarelli F, Mellors JW. 2012. Short-course raltegravir intensification does not increase 2 long terminal repeat episomal HIV-1 DNA in patients on effective antiretroviral therapy. Clin Infect Dis 54:451–453. doi:10.1093/cid/cir721.
    1. Kearney MF, Spindler J, Shao W, Yu S, Anderson EM, O’Shea A, Rehm C, Poethke C, Kovacs N, Mellors JW, Coffin JM, Maldarelli F. 2014. Lack of detectable HIV-1 molecular evolution during suppressive antiretroviral therapy. PLoS Pathog 10:e1004010. doi:10.1371/journal.ppat.1004010.
    1. Deeks SG, Lewin SR, Havlir DV. 2013. The end of AIDS: HIV infection as a chronic disease. Lancet 382:1525–1533. doi:10.1016/S0140-6736(13)61809-7.
    1. Dinoso JB, Kim SY, Wiegand AM, Palmer SE, Gange SJ, Cranmer L, O’Shea A, Callender M, Spivak A, Brennan T, Kearney MF, Proschan MA, Mican JM, Rehm CA, Coffin JM, Mellors JW, Siliciano RF, Maldarelli F. 2009. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci U S A 106:9403–9408. doi:10.1073/pnas.0903107106.
    1. Gandhi RT, Zheng L, Bosch RJ, Chan ES, Margolis DM, Read S, Kallungal B, Palmer S, Medvik K, Lederman MM, Alatrakchi N, Jacobson JM, Wiegand A, Kearney M, Coffin JM, Mellors JW, Eron JJ, AIDS Clinical Trials Group A5244 Team . 2010. The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med 7:e1000321. doi:10.1371/journal.pmed.1000321.
    1. Okamoto T, Wong-Staal F. 1986. Demonstration of virus-specific transcriptional activator(s) in cells infected with HTLV-III by an in vitro cell-free system. Cell 47:29–35. doi:10.1016/0092-8674(86)90363-6.
    1. Selby MJ, Bain ES, Luciw PA, Peterlin BM. 1989. Structure, sequence, and position of the stem-loop in TAR determine transcriptional elongation by tat through the HIV-1 long terminal repeat. Genes Dev 3:547–558. doi:10.1101/gad.3.4.547.
    1. Laspia MF, Rice AP, Mathews MB. 1989. HIV-1 Tat protein increases transcriptional initiation and stabilizes elongation. Cell 59:283–292. doi:10.1016/0092-8674(89)90290-0.
    1. Mancebo HS, Lee G, Flygare J, Tomassini J, Luu P, Zhu Y, Peng J, Blau C, Hazuda D, Price D, Flores O. 1997. P-TEFb kinase is required for HIV Tat transcriptional activation in vivo and in vitro. Genes Dev 11:2633–2644. doi:10.1101/gad.11.20.2633.
    1. Wei P, Garber ME, Fang SM, Fischer WH, Jones KA. 1998. A novel CDK9-associated C-type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA. Cell 92:451–462. doi:10.1016/S0092-8674(00)80939-3.
    1. Karn J. 2011. The molecular biology of HIV latency: breaking and restoring the Tat-dependent transcriptional circuit. Curr Opin HIV AIDS 6:4–11. doi:10.1097/COH.0b013e328340ffbb.
    1. Ott M, Geyer M, Zhou Q. 2011. The control of HIV transcription: keeping RNA polymerase II on track. Cell Host Microbe 10:426–435. doi:10.1016/j.chom.2011.11.002.
    1. Ghose R, Liou LY, Herrmann CH, Rice AP. 2001. Induction of TAK (cyclin T1/P-TEFb) in purified resting CD4(+) T lymphocytes by combination of cytokines. J Virol 75:11336–11343. doi:10.1128/JVI.75.23.11336-11343.2001.
    1. Kinoshita S, Chen BK, Kaneshima H, Nolan GP. 1998. Host control of HIV-1 parasitism in T cells by the nuclear factor of activated T cells. Cell 95:595–604. doi:10.1016/S0092-8674(00)81630-X.
    1. Nabel G, Baltimore D. 1987. An inducible transcription factor activates expression of human immunodeficiency virus in T cells. Nature 326:711–713. doi:10.1038/326711a0.
    1. Tyagi M, Karn J. 2007. CBF-1 promotes transcriptional silencing during the establishment of HIV-1 latency. EMBO J 26:4985–4995. doi:10.1038/sj.emboj.7601928.
    1. He G, Margolis DM. 2002. Counterregulation of chromatin deacetylation and histone deacetylase occupancy at the integrated promoter of human immunodeficiency virus type 1 (HIV-1) by the HIV-1 repressor YY1 and HIV-1 activator Tat. Mol Cell Biol 22:2965–2973. doi:10.1128/MCB.22.9.2965-2973.2002.
    1. Baldauf HM, Pan X, Erikson E, Schmidt S, Daddacha W, Burggraf M, Schenkova K, Ambiel I, Wabnitz G, Gramberg T, Panitz S, Flory E, Landau NR, Sertel S, Rutsch F, Lasitschka F, Kim B, König R, Fackler OT, Keppler OT. 2012. SAMHD1 restricts HIV-1 infection in resting CD4(+) T cells. Nat Med 18:1682–1687. doi:10.1038/nm.2964.
    1. Gao WY, Cara A, Gallo RC, Lori F. 1993. Low levels of deoxynucleotides in peripheral blood lymphocytes: a strategy to inhibit human immunodeficiency virus type 1 replication. Proc Natl Acad Sci U S A 90:8925–8928. doi:10.1073/pnas.90.19.8925.
    1. Lenasi T, Contreras X, Peterlin BM. 2008. Transcriptional interference antagonizes proviral gene expression to promote HIV latency. Cell Host Microbe 4:123–133. doi:10.1016/j.chom.2008.05.016.
    1. Van Lint C. 2000. Role of chromatin in HIV-1 transcriptional regulation. Adv Pharmacol 48:121–160. doi:10.1016/S1054-3589(00)48005-1.
    1. Van Lint C, Emiliani S, Ott M, Verdin E. 1996. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. EMBO J 15:1112–1120.
    1. Lusic M, Giacca M. 2015. Regulation of HIV-1 latency by chromatin structure and nuclear architecture. J Mol Biol 427:688–694. doi:10.1016/j.jmb.2014.07.022.
    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. 2009. CpG methylation controls reactivation of HIV from latency. PLoS Pathog 5:e1000554. doi:10.1371/journal.ppat.1000554.
    1. Blazkova J, Murray D, Justement JS, Funk EK, Nelson AK, Moir S, Chun TW, Fauci AS. 2012. Paucity of HIV DNA methylation in latently infected, resting CD4+ T cells from infected individuals receiving antiretroviral therapy. J Virol 86:5390–5392. doi:10.1128/JVI.00040-12.
    1. Ho Y-C, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DI, Lai J, Blankson JN, Siliciano JD, Siliciano RF. 2013. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 155:540–551. doi:10.1016/j.cell.2013.09.020.
    1. Mahmoudi T, Parra M, Vries RG, Kauder SE, Verrijzer CP, Ott M, Verdin E. 2006. The SWI/SNF chromatin-remodeling complex is a cofactor for Tat transactivation of the HIV promoter. J Biol Chem 281:19960–19968. doi:10.1074/jbc.M603336200.
    1. Tréand C, du Chéné I, Brès V, Kiernan R, Benarous R, Benkirane M, Emiliani S. 2006. Requirement for SWI/SNF chromatin-remodeling complex in Tat-mediated activation of the HIV-1 promoter. EMBO J 25:1690–1699. doi:10.1038/sj.emboj.7601074.
    1. Gerritsen ME, Williams AJ, Neish AS, Moore S, Shi Y, Collins T. 1997. CREB-binding protein/p300 are transcriptional coactivators of p65. Proc Natl Acad Sci U S A 94:2927–2932. doi:10.1073/pnas.94.7.2927.
    1. Agbottah E, Deng L, Dannenberg LO, Pumfery A, Kashanchi F. 2006. Effect of SWI/SNF chromatin remodeling complex on HIV-1 Tat activated transcription. Retrovirology 3:48. doi:10.1186/1742-4690-3-48.
    1. Benkirane M, Chun RF, Xiao H, Ogryzko VV, Howard BH, Nakatani Y, Jeang KT. 1998. Activation of integrated provirus requires histone acetyltransferase. p300 and P/CAF are coactivators for HIV-1 Tat. J Biol Chem 273:24898–24905. doi:10.1074/jbc.273.38.24898.
    1. Col E, Caron C, Seigneurin-Berny D, Gracia J, Favier A, Khochbin S. 2001. The histone acetyltransferase, hGCN5, interacts with and acetylates the HIV transactivator, Tat. J Biol Chem 276:28179–28184. doi:10.1074/jbc.M101385200.
    1. Hottiger MO, Nabel GJ. 1998. Interaction of human immunodeficiency virus type 1 Tat with the transcriptional coactivators p300 and CREB binding protein. J Virol 72:8252–8256.
    1. Marzio G, Tyagi M, Gutierrez MI, Giacca M. 1998. HIV-1 tat transactivator recruits p300 and CREB-binding protein histone acetyltransferases to the viral promoter. Proc Natl Acad Sci U S A 95:13519–13524. doi:10.1073/pnas.95.23.13519.
    1. Demarchi F, Gutierrez MI, Giacca M. 1999. Human immunodeficiency virus type 1 tat protein activates transcription factor NF-κB through the cellular interferon-inducible, double-stranded RNA-dependent protein kinase, PKR. J Virol 73:7080–7086.
    1. Razooky BS, Pai A, Aull K, Rouzine IM, Weinberger LS. 2015. A hardwired HIV latency program. Cell 160:990–1001. doi:10.1016/j.cell.2015.02.009.
    1. Yukl S, Pillai S, Li P, Chang K, Pasutti W, Ahlgren C, Havlir D, Strain M, Günthard H, Richman D, Rice AP, Daar E, Little S, Wong JK. 2009. Latently infected CD4+ T cells are enriched for HIV-1 Tat variants with impaired transactivation activity. Virology 387:98–108. doi:10.1016/j.virol.2009.01.013.
    1. Mousseau G, Mediouni S, Valente ST. 2015. Targeting HIV transcription: the quest for a functional cure. Curr Top Microbiol Immunol 389:121–145 doi:10.1007/82_2015_435.
    1. Mousseau G, Clementz MA, Bakeman WN, Nagarsheth N, Cameron M, Shi J, Baran P, Fromentin R, Chomont N, Valente ST. 2012. An analog of the natural steroidal alkaloid cortistatin A potently suppresses Tat-dependent HIV transcription. Cell Host Microbe 12:97–108. doi:10.1016/j.chom.2012.05.016.
    1. Pasternak AO, Adema KW, Bakker M, Jurriaans S, Berkhout B, Cornelissen M, Lukashov VV. 2008. Highly sensitive methods based on seminested real-time reverse transcription-PCR for quantitation of human immunodeficiency virus type 1 unspliced and multiply spliced RNA and proviral DNA. J Clin Microbiol 46:2206–2211. doi:10.1128/JCM.00055-08.
    1. Chun TW, Engel D, Mizell SB, Ehler LA, Fauci AS. 1998. Induction of HIV-1 replication in latently infected CD4+ T cells using a combination of cytokines. J Exp Med 188:83–91. doi:10.1084/jem.188.1.83.
    1. Scripture-Adams DD, Brooks DG, Korin YD, Zack JA. 2002. Interleukin-7 induces expression of latent human immunodeficiency virus type 1 with minimal effects on T-cell phenotype. J Virol 76:13077–13082. doi:10.1128/JVI.76.24.13077-13082.2002.
    1. Wang FX, Xu Y, Sullivan J, Souder E, Argyris EG, Acheampong EA, Fisher J, Sierra M, Thomson MM, Najera R, Frank I, Kulkosky J, Pomerantz RJ, Nunnari G. 2005. IL-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 115:128–137. doi:10.1172/JCI22574.
    1. Chan JK, Greene WC. 2012. Dynamic roles for NF-κB in HTLV-I and HIV-1 retroviral pathogenesis. Immunol Rev 246:286–310. doi:10.1111/j.1600-065X.2012.01094.x.
    1. Pessler F, Cron RQ. 2004. Reciprocal regulation of the nuclear factor of activated T cells and HIV-1. Genes Immun 5:158–167. doi:10.1038/sj.gene.6364047.
    1. Spina CA, Anderson J, Archin NM, Bosque A, Chan J, Famiglietti M, Greene WC, Kashuba A, Lewin SR, Margolis DM, Mau M, Ruelas D, Saleh S, Shirakawa K, Siliciano RF, Singhania A, Soto PC, Terry VH, Verdin E, Woelk C, Wooden S, Xing S, Planelles V. 2013. An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog 9:e1003834. doi:10.1371/journal.ppat.1003834.
    1. Mbonye U, Karn J. 2011. Control of HIV latency by epigenetic and non-epigenetic mechanisms. Curr HIV Res 9:554–567. doi:10.2174/157016211798998736.
    1. Weinberger LS, Burnett JC, Toettcher JE, Arkin AP, Schaffer DV. 2005. Stochastic gene expression in a lentiviral positive-feedback loop: HIV-1 Tat fluctuations drive phenotypic diversity. Cell 122:169–182. doi:10.1016/j.cell.2005.06.006.
    1. Hocqueloux L, Prazuck T, Avettand-Fenoel V, Lafeuillade A, Cardon B, Viard JP, Rouzioux C. 2010. Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. AIDS 24:1598–1601. doi:10.1097/QAD.0b013e32833b61ba.
    1. Butera ST, Perez VL, Wu BY, Nabel GJ, Folks TM. 1991. Oscillation of the human immunodeficiency virus surface receptor is regulated by the state of viral activation in a CD4+ cell model of chronic infection. J Virol 65:4645–4653.
    1. Jordan A, Bisgrove D, Verdin E. 2003. HIV reproducibly establishes a latent infection after acute infection of T cells in vitro. EMBO J 22:1868–1877. doi:10.1093/emboj/cdg188.
    1. Chávez L, Kauder S, Verdin E. 2011. In vivo, in vitro, and in silico analysis of methylation of the HIV-1 provirus. Methods 53:47–53. doi:10.1016/j.ymeth.2010.05.009.
    1. Kauder SE, Bosque A, Lindqvist A, Planelles V, Verdin E. 2009. Epigenetic regulation of HIV-1 latency by cytosine methylation. PLoS Pathog 5:e1000495. doi:10.1371/journal.ppat.1000495.
    1. Chatila T, Silverman L, Miller R, Geha R. 1989. Mechanisms of T cell activation by the calcium ionophore ionomycin. J Immunol 143:1283–1289.
    1. Siekevitz M, Josephs SF, Dukovich M, Peffer N, Wong-Staal F, Greene WC. 1987. Activation of the HIV-1 LTR by T cell mitogens and the trans-activator protein of HTLV-I. Science 238:1575–1578. doi:10.1126/science.2825351.
    1. Hiscott J, Marois J, Garoufalis J, D’Addario M, Roulston A, Kwan I, Pepin N, Lacoste J, Nguyen H, Bensi G, et al. . 1993. Characterization of a functional NF-kappa B site in the human interleukin 1β promoter: evidence for a positive autoregulatory loop. Mol Cell Biol 13:6231–6240.
    1. Kolch W, Heidecker G, Kochs G, Hummel R, Vahidi H, Mischak H, Finkenzeller G, Marmé D, Rapp UR. 1993. Protein kinase C alpha activates RAF-1 by direct phosphorylation. Nature 364:249–252. doi:10.1038/364249a0.
    1. Schneidewind A, Brockman MA, Yang R, Adam RI, Li B, Le Gall S, Rinaldo CR, Craggs SL, Allgaier RL, Power KA, Kuntzen T, Tung CS, LaBute MX, Mueller SM, Harrer T, McMichael AJ, Goulder PJ, Aiken C, Brander C, Kelleher AD, Allen TM. 2007. Escape from the dominant HLA-B27-restricted cytotoxic T-lymphocyte response in gag is associated with a dramatic reduction in human immunodeficiency virus type 1 replication. J Virol 81:12382–12393. doi:10.1128/JVI.01543-07.
    1. McKernan LN, Momjian D, Kulkosky J. 2012. Protein kinase C: one pathway towards the eradication of latent HIV-1 reservoirs. Adv Virol 2012:805347. doi:10.1155/2012/805347.
    1. Williams SA, Chen LF, Kwon H, Fenard D, Bisgrove D, Verdin E, Greene WC. 2004. Prostratin antagonizes HIV latency by activating NF-κB. J Biol Chem 279:42008–42017. doi:10.1074/jbc.M402124200.
    1. Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, Margolis DM. 2009. Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Hum Retroviruses 25:207–212. doi:10.1089/aid.2008.0191.
    1. Clouse KA, Powell D, Washington I, Poli G, Strebel K, Farrar W, Barstad P, Kovacs J, Fauci AS, Folks TM. 1989. Monokine regulation of human immunodeficiency virus-1 expression in a chronically infected human T cell clone. J Immunol 142:431–438.
    1. Folks TM, Clouse KA, Justement J, Rabson A, Duh E, Kehrl JH, Fauci AS. 1989. Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci U S A 86:2365–2368. doi:10.1073/pnas.86.7.2365.
    1. Folks TM, Justement J, Kinter A, Dinarello CA, Fauci AS. 1987. Cytokine-induced expression of HIV-1 in a chronically infected promonocyte cell line. Science 238:800–802. doi:10.1126/science.3313729.
    1. Cannon P, Kim SH, Ulich C, Kim S. 1994. Analysis of Tat function in human immunodeficiency virus type 1-infected low-level-expression cell lines U1 and ACH-2. J Virol 68:1993–1997.
    1. Emiliani S, Van Lint C, Fischle W, Paras P Jr., Ott M, Brady J, Verdin E. 1996. A point mutation in the HIV-1 Tat responsive element is associated with postintegration latency. Proc Natl Acad Sci U S A 93:6377–6381. doi:10.1073/pnas.93.13.6377.
    1. Emiliani S, Fischle W, Ott M, Van Lint C, Amella CA, Verdin E. 1998. Mutations in the tat gene are responsible for human immunodeficiency virus type 1 postintegration latency in the U1 cell line. J Virol 72:1666–1670.
    1. Reza SM, Rosetti M, Mathews MB, Pe’ery T. 2003. Differential activation of Tat variants in mitogen-stimulated cells: implications for HIV-1 postintegration latency. Virology 310:141–156. doi:10.1016/S0042-6822(03)00106-5.
    1. Adams M, Sharmeen L, Kimpton J, Romeo JM, Garcia JV, Peterlin BM, Groudine M, Emerman M. 1994. Cellular latency in human immunodeficiency virus-infected individuals with high CD4 levels can be detected by the presence of promoter-proximal transcripts. Proc Natl Acad Sci U S A 91:3862–3866. doi:10.1073/pnas.91.9.3862.
    1. Adams M, Wong C, Wang D, Romeo J. 1999. Limitation of Tat-associated transcriptional processivity in HIV-infected PBMC. Virology 257:397–405. doi:10.1006/viro.1999.9647.
    1. Ambrosino C, Ruocco MR, Chen X, Mallardo M, Baudi F, Trematerra S, Quinto I, Venuta S, Scala G. 1997. HIV-1 Tat induces the expression of the interleukin-6 (IL6) gene by binding to the IL6 leader RNA and by interacting with CAAT enhancer-binding protein beta (NF-IL6) transcription factors. J Biol Chem 272:14883–14892. doi:10.1074/jbc.272.23.14883.
    1. Chen R, Liu M, Li H, Xue Y, Ramey WN, He N, Ai N, Luo H, Zhu Y, Zhou N, Zhou Q. 2008. PP2B and PP1α cooperatively disrupt 7SK snRNP to release P-TEFb for transcription in response to Ca2+ signaling. Genes Dev 22:1356–1368. doi:10.1101/gad.1636008.
    1. Chun RF, Semmes OJ, Neuveut C, Jeang KT. 1998. Modulation of Sp1 phosphorylation by human immunodeficiency virus type 1 Tat. J Virol 72:2615–2629.
    1. Verhoef K, Koper M, Berkhout B. 1997. Determination of the minimal amount of Tat activity required for human immunodeficiency virus type 1 replication. Virology 237:228–236. doi:10.1006/viro.1997.8786.
    1. Meyerhans A, Cheynier R, Albert J, Seth M, Kwok S, Sninsky J, Morfeldt-Månson L, Asjö B, Wain-Hobson S. 1989. Temporal fluctuations in HIV quasispecies in vivo are not reflected by sequential HIV isolations. Cell 58:901–910. doi:10.1016/0092-8674(89)90942-2.
    1. Sabino E, Cheng-Mayer C, Mayer A. 1993. An individual with a high prevalence of a tat-defective provirus in peripheral blood. AIDS Res Hum Retroviruses 9:1265–1268. doi:10.1089/aid.1993.9.1265.
    1. Donahue DA, Kuhl BD, Sloan RD, Wainberg MA. 2012. The viral protein Tat can inhibit the establishment of HIV-1 latency. J Virol 86:3253–3263. doi:10.1128/JVI.06648-11.
    1. Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, Parker DC, Anderson EM, Kearney MF, Strain MC, Richman DD, Hudgens MG, Bosch RJ, Coffin JM, Eron JJ, Hazuda DJ, Margolis DM. 2012. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487:482–485. doi:10.1038/nature11286.
    1. Deeks SG. 2012. HIV: shock and kill. Nature 487:439–440. doi:10.1038/487439a.
    1. Shan L, Deng K, Shroff NS, Durand CM, Rabi SA, Yang HC, Zhang H, Margolick JB, Blankson JN, Siliciano RF. 2012. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity 36:491–501. doi:10.1016/j.immuni.2012.01.014.
    1. Elliott JH, Wightman F, Solomon A, Ghneim K, Ahlers J, Cameron MJ, Smith MZ, Spelman T, McMahon J, Velayudham P, Brown G, Roney J, Watson J, Prince MH, Hoy JF, Chomont N, Fromentin R, Procopio FA, Zeidan J, Palmer S, Odevall L, Johnstone RW, Martin BP, Sinclair E, Deeks SG, Hazuda DJ, Cameron PU, Sekaly RP, Lewin SR. 2014. Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS Pathog 10:e1004473. doi:10.1371/journal.ppat.1004473.
    1. Ramratnam B, Mittler JE, Zhang L, Boden D, Hurley A, Fang F, Macken CA, Perelson AS, Markowitz M, Ho DD. 2000. The decay of the latent reservoir of replication-competent HIV-1 is inversely correlated with the extent of residual viral replication during prolonged anti-retroviral therapy. Nat Med 6:82–85. doi:10.1038/71577.

Source: PubMed

3
Předplatit