Skip to main content

Cell line-dependent variability in HIV activation employing DNMT inhibitors

Abstract

Long-lived reservoirs of Human Immunodeficiency Virus (HIV) latently infected cells present the main barrier to a cure for HIV infection. Much interest has focused on identifying strategies to activate HIV, which would be used together with antiretrovirals to attack reservoirs. Several HIV activating agents, including Tumor Necrosis Factor alpha (TNFα) and other agents that activate via NF-kB are not fully effective in all latent infection models due to epigenetic restrictions, such as DNA methylation and the state of histone acetylation. DNA methyltransferases (DNMT) inhibitors like 5-aza-2'deoxycytidine (Aza-CdR) and histone deacetylase (HDAC) inhibitors like Trichostatin A (TSA) have been proposed as agents to enhance reactivation and have shown activity in model systems. However, it is not clear how the activities of DNMT and HDAC inhibitors range across different latently infected cell lines, potential models for the many different latently infected cells within an HIV patient. We determined HIV activation following treatment with TNFα, TSA and Aza-CdR across a range of well known latently infected cell lines. We assessed the activity of these compounds in four different Jurkat T cell-derived J-Lat cell lines (6.3, 8.4, 9.2 and 10.6), which have a latent HIV provirus in which GFP replaces Nef coding sequence, and ACH-2 and J1.1 (T cell-derived), and U1 (promonocyte-derived) cell lines with full-length provirus. We found that Aza-CdR plus TNFα activated HIV at least twice as well as TNFα alone for almost all J-Lat cells, as previously described, but not for J-Lat 10.6, in which TNFα plus Aza-CdR moderately decreased activation compared to TNFα alone. Surprisingly, a much greater reduction of TNFα-stimulated activation with Aza-CdR was detected for ACH-2, J1.1 and U1 cells. Reaching the highest reduction in U1 cells with a 75% reduction. Interestingly, Aza-CdR not only decreased TNFα induction of HIV expression in certain cell lines, but also decreased activation by TSA. Since DNMT inhibitors reduce the activity of provirus activators in some HIV latently infected cell lines the use of epigenetic modifying agents may need to be carefully optimized if they are to find clinical utility in therapies aimed at attacking latent HIV reservoirs.

Findings

Despite the effectiveness of Highly Active Antiretroviral Therapy (HAART) for Human Immunodeficiency Virus type 1 (HIV-1) infection, patients cannot be cured due to the persistence of long-lived reservoirs of cells latently infected with HIV ([1–5] and reviewed in [6–11]). Much interest has focused on attacking this reservoir of HIV latently infected cells. A potentially useful strategy, sometimes termed "shock and kill" [12], aims to attack the latent reservoir treating patients with HIV-activating agents to stimulate HIV replication in the latently infected cells while blocking new infection of cells with antiretrovirals.

One activation approach employs agents like phorbol esters (e.g 12-O-Tetradecanoylphorbol-13-acetate (TPA), prostratin) [13], interleukins (IL-2, IL-7) [14–18] and cytokines (e.g. tumor necrosis factor alpha (TNFα)) [19] that directly activate HIV gene expression via well-known transcriptional activation pathways, like NF-κB, but many such agents are toxic or incompletely effective in vivo [14, 16, 20, 21]. Other approaches target the provirus' epigenetic environment, employing histone deacetylase (HDAC) inhibitors (HDACIs) like trichostatin A (TSA) [22, 23], suberoylanilide hydroxamic acid (SAHA) [22, 24], sodium butyrate [25, 26], and valproic acid [27, 28], and/or DNA methyltransferase (DNMT) inhibitors (DNMTIs) like 5-aza-2'deoxycytidine (Aza-CdR) [29], with some strategies combining approaches [30] (and reviewed in [6, 31]). In addition, to increase the efficiency of viral activators, combining the use of activators with compounds aimed at limiting the toxicity of the activator, as shown with buthionine sulfoximine [32] may play a major role in optimizing treatment strategies.

Nucleosomes occupy specific positions on the HIV LTR [22, 33, 34], with the nucleosome occupying the position termed 'nuc-1' at a regulatory region of the LTR 610 to 720 bp 3' to the transcription start site having significant inhibitory effects on HIV expression. Chromatin condensed around nuc-1 in its deacetylated form presents a block to HIV expression. Following hyperacetylation and chromatin remodeling, mediated by the recruitment of histone acetylases by transcription factors binding directly or indirectly to the LTR, or by Tat, the nuc-1 block is removed [22, 33, 35–37]. HDACIs, by shifting histones to a more acetylated state facilitate remodeling and removal of the block [38, 39]. DNMTIs likely activate HIV because, at least in some cells, the LTR contains two CpG islands (particularly island 2) that can be hypermethylated and the hypermethylated DNA can recruit methyl-CpG binding domain (MDB) protein family members, notably MDB2. MDB2 recruited to the LTR can serve a bridging function between DNA and chromatin-modifying factors, such as HDACs [29]. Several studies linked retroviral promoter CpG methylation with transcription inactivation [40–42] and cellular gene silencing [43]. DNMTIs can moderately activate HIV alone, but in some systems they significantly enhance agents, like TNFα, that directly activate the HIV LTR, probably because activation is limited due to proviral DNA hypermethylation [29, 44, 45].

While DNMTIs and HDACIs significantly enhance HIV activation in some latently infected cells, their ability to enhance activation across a wider range of latently infected cells is currently unknown. The breadth of activating ability is important, since complete or close-to complete eradication of latently infected cells may be required to cure HIV infection. To better assess the breadth of activity of activators representative of both direct activators and activators acting via epigenetic effects we surveyed the ability of three prototypical HIV activating agents, the DNMTI Aza-CdR (2.5 μM), the HDACI TSA (1.5 μM), and TNFα (10 ng/ml), activating via NF-κB, in a panel of widely used HIV latently infected model cell lines, including the J-Lat series derived from Jurkat T-cells [46], J1.1 [47], also a Jurkat derivative, ACH-2 [48], derived from the A3.01 T lymphocytoid cell line, and U1 [49], derived from the U937 promonocytic cell line. The concentrations used for TSA and TNFα were optimized to obtain the best activation profile in combination with Aza-CdR. A dose response curve with J-Lat 6.3 cells was performed to determine the minimum concentration required where the activation reached a plateau (data not shown). We employed this plateau concentration in subsequent studies to assess the effects of combined treatments with different agents used at their maximally effective concentrations, since the object of any latency activation strategy would be to determine how to achieve a maximal degree of provirus activation.

J-Lat cells harbor an HIV provirus containing the Green Fluorescent Protein (GFP) ORF instead of nef and a frameshift mutation in env [46]. With GFP under the control of the HIV LTR, activation is conveniently monitored via flow cytometry. HIV production in J-Lat cells can also be determined by p24 antigen (p24) quantification. Different J-Lat lines exhibit different activation patterns when treated with TNFα, likely due to different integration sites and accompanying epigenetic states [46]. J-Lat cells have contributed significantly to latency studies, including the identification of the methyl-CpG binding domain protein 2 (MBD2), a transcriptional repressor that binds to methylated DNA, as a regulator of HIV latency, which suggested that DNMTIs like Aza-CdR might activate latent provirus [29]. Moreover, LTR demethylation, after Aza-CdR treatment, correlated with a synergistic increase of proviral activation when combined with TNFα in J-Lat 6.3, 8.4, 9.2 and 15.4. ACH-2, U1 and J1.1 harbor full-length proviruses, without GFP replacing coding sequence (although J1.1 has defects in signalling through CD3, ACH-2 has a point mutation in the Tat responsive element (TAR) [50], and U1 contains mutations in Tat [51]). Flow cytometric evaluation of activation is not possible in these lines, but they can be monitored using assays for HIV gene products (e.g. p24).

Before embarking on a survey of the activators across cell lines, we undertook further optimization experiments, initially performing order-of-addition experiments using J-Lat 6.3 (Fig.1), in which Aza-CdR had been shown to enhance activation mediated by TNFα [29], since inhibiting DNA methylation well before addition of the direct transcriptional activator TNFα could be required for maximal activation. Detection and quantification of GFP positive cells from a live-gated population was performed at 48 h using a FACScalibur cytometer (BD Biosciences, San Jose CA), analyzed with Cell Quest Pro software (BD Biosciences, San Jose CA). As has been previously reported, we found that Aza-CdR alone did not activate [29], but TNFα alone produced significant increases in GFP positive cells, and Aza-CdR enhanced activation with TNFα treatment. We calculated the fold increase of activation due to Aza-CdR, that is the activation observed with TNFα plus Aza-CdR divided by the activation with TNFα alone (TNFα+Aza-CdR/TNFα, (FI-Aza)), which expresses the fold increase due to the addition of Aza-CdR. FI-Aza showed that the greatest activation occurred when J-Lat 6.3 cells were treated with both compounds simultaneously (FI-Aza:3.8). Although there were differences in the extent of activation that depended on the order of addition, Aza-CdR always enhanced activation of J-Lat 6.3 by TNFα, with FI-Aza ranging between 3.8 and 1.9.

Figure 1
figure 1

Optimization of J-Lat 6.3 cells activation by TNFα/Aza-CdR. HIV latently infected J-Lat 6.3 cells that contain the GFP reporter gene were (A) treated with Aza-CdR (2.5 μM) and TNFα (10 ng/ml) for 48 h, (B) were treated with Aza-CdR plus TNFα and then washed at 24 h, (C) treated with Aza-CdR followed by addition of TNFα after 24 h of Aza-CdR treatment, and (D) treated with Aza-CdR plus TNFα for 24 h, followed by the addition of fresh Aza-CdR. Cells were cultured in RPMI (Atlanta Biologicals, Lawrenceville, GA) with 10% FBS (Invitrogen, Carlsbad, CA). Two million cells were seeded with 3 ml medium in six well plates and treated with Aza-CdR (2.5 μM), TNFα (10 ng/ml) or both compounds combined. After 48 h cells were transferred to a 15 ml conical tube and spun down at 1400 rpm for 7 min. The supernatant was discarded and cell pellets were resuspended and were washed twice with chilled 1× PBS and fixed in 2% formaldehyde for FACS analysis using a FACScalibur cytometer (BD Biosciences, San Jose CA) and analyzed with Cell Quest Pro software (BD Biosciences, San Jose CA). GFP positive cells from the live population, defined by forward versus side scatter gating, were quantified. 20.000 events per treatment condition were analyzed. Results are the mean ± standard deviation (SD). The TNFαAza-CdR/TNFα (FI-Aza) ratio was calculated and used to evaluate the optimum time of activator addition.

To determine when Aza-CdR most enhanced TNFα or TSA activation, a time-course experiment was performed using J-Lat 6.3 cells. Percent GFP positive cells using FACS analysis (Fig. 2A) and p24 production using an enzyme-linked immunosorbent assay (ELISA) (Perkin Elmer, Waltham, MA) (Fig. 2B) were determined for all time points and treatment conditions. Aza-CdR in combination with TNFα reached its maximum effect at 48 h post-induction when per cent GFP positive cells where quantified. Moreover, p24 production did not increase significantly beyond 48 h, losing Aza-CdR's enhancing effect when combined with TNFα at later time points. Due to the lack of synergy between TSA and Aza-CdR we determined 48 h to be the best time point to analyze further data based on TNFα plus Aza-CdR results. Cell viability for all time points and treatment conditions (Fig. 2C) were performed using MTS assay (Promega, Madison, WI). TNFα alone does not strongly activate J-Lat 6.3, 8.4 and 9.2 cells, as shown in Fig. 3A, and as reported [29]. TNFα can, at most, achieve 20% activation in these three cell lines. In contrast, in J-Lat 10.6, TNFα activates ~80% of the cells, which enabled us to determine the effect of combining Aza-CdR with TNFα and TSA when the provirus is highly activated by TNFα. Surprisingly, the combination of Aza-CdR and TNFα did not increase or maintain activation in J-Lat 10.6, but instead moderately decreased activation (p ≤ 0.1) (Fig. 3B) in per cent GFP positive cells while activation from the combination of Aza-CdR and TNFα was significantly lower when p24 was determined (p ≤ 0.05) (Fig. 3C). The reduction of activation in J-Lat 10.6 suggested that Aza-CdR could have, in some latently infected cells, a detrimental effect when combined with an activator, particularly when activation via other pathways, such as those stimulated by TNFα, is great (Fig. 3C). Cell viability did not differ between TNFα and Aza-CdR, alone or in combination (Fig. 3D), so cell toxicity could not account for the observations. Of note, we did not detect a synergistic effect when TNFα and TSA were combined when percent GFP positive cells were quantified. As shown in the time-course experiment in Fig. 2 this synergy can be detected in earlier time points but not at 48 h post-induction when Aza-CdR reaches maximal effect. When p24 production is determined, we only could see this TSA-TNFα synergy in J-Lat 8.4 cells because TNFα activation in this cell line is strongly blocked (Fig. 3C).

Figure 2
figure 2

Time-course activation by TNFα and TSA alone or in combination with Aza-CdR in J-Lat 6.3 cells. A. GFP positive cell quantification by FACS analysis at 24, 48 and 72 h post-induction for all treatments and treatment combinations using J-Lat 6.3 cell line. GFP positive cells from the live population, defined by forward versus side scatter gating, were quantified. 20.000 events per treatment condition were analyzed. B. p24 antigen production determined using an enzyme-linked immunosorbent assay (ELISA) (Perkin Elmer, Waltham, MA) for all post-induction time points and treatments as for panel A. C. Cell viability of the different treatments and treatment combinations for all time points determined with MTS assay (Promega, Madison, WI). Final activator compound concentrations were, for Aza-CdR (2.5 μM), TNFα (10 ng/ml) and TSA (1.5 μM). Results are the mean ± standard deviation (SD).

Figure 3
figure 3

HIV activation by TNFα and TSA alone or in combination with Aza-CdR in four J-Lat cell lines. A. GFP positive cell quantification by FACS analysis at 48 h post-induction of J-Lat 6.3, 8.4, 9.2 and 10.6 cell lines treated with TNFα or Aza-CdR alone or in combination. 20.000 events from the live population defined by forward versus side scatter gating were analyzed. B. Proviral activation as determined by percent GFP positive cells for the different treatments and treatment combinations in the four J-Lat cells (J-Lat 6.3, 8.4, 9.2 and 10.6 indicated on each panel). C. p24 antigen production for all treatments and treatment combinations at 48 h post-induction. D. Cell viability of the different treatments and treatment combinations determined with an MTS assay (Promega, Madison, WI) at 48 h. Cells were cultured in RPMI with 10% FBS. Two million cells were seeded and treated with the different compounds for 48 h. The compound order of addition was determined as described in Fig. 1: agents were added at time 0 h, with no further additional steps. After 48 h, cells were washed twice with chilled 1× PBS and fixed in 2% formaldehyde as in the FACS analysis described in the Fig. 1 legend. At the same time, supernatants from the different treatment conditions were stored at -80°C until further use. p24 antigen was determined using ELISA (Perkin Elmer, Waltham, MA) from the stored supernatants. Final activator compound concentrations were, for Aza-CdR (2.5 μM), TNFα (10 ng/ml) and TSA (1.5 μM). Results are the mean ± standard deviation (SD). Statistical analysis (Student's t-test) was performed using the STATA software package (StataCorp LP, College Station, TX), * p ≤ 0.05.

To assess how the combinations of Aza-CdR, TNFα, and TSA affect HIV activation in other HIV latently infected cells, we studied the ability of these agents, alone and together, to activate HIV in ACH-2, U1 and J1.1 cells (Fig. 4A), other lines in which TNFα strongly activates HIV expression [52]. Since these cells harbor a latent HIV provirus that does not have nef replaced with GFP, we assessed activation by measuring p24 48 h after treatment with the activators using ELISA (Perkin Elmer, Waltham, MA). TNFα powerfully activated HIV expression in these cells, as previously described, but as we observed with J-Lat 10.6, treating ACH-2, U1 and J1.1 cells with Aza-CdR significantly decreased activation by TNFα, from 23% (J1.1, p ≤ 0.05) to 40% (ACH-2, p ≤ 0.05), to 75% (U1, p ≤ 0.05). In ACH-2 cells, but not in any other of the cell lines studied, Aza-CdR also significantly decreased activation following treatment with TSA alone (57%, p ≤ 0.05), or TSA+TNFα (45%, p ≤ 0.05). Impaired cell viability, as with J-Lat cells, could not explain this reduction in activation (Fig. 4B). Thus, in certain circumstances, a DNMTI can decrease activation by agents acting both through the NF-κB pathway and mediated by an HDACI. However, a decrease in activation produced by Aza-CdR was not observed in all the non-J-Lat cell lines under all conditions. In J1.1 cells, Aza-CdR did not decrease activation by TSA or TSA plus TNFα. Overall, the ability of the DNMTI Aza-CdR to help activate HIV replication (or to inhibit the activation produced by other agents) exhibited a strong cell line dependence: In U1 and J1.1 cell lines, Aza-CdR inhibited activation by TNFα, but not by TSA or TNFα plus TSA, while in ACH-2 cells Aza-CdR inhibited activation by both TNFα and TSA when used alone or in combination. Table 1 summarizes the effect of Aza-CdR and TNFα when added simultaneously in all latently infected cells tested, highlighting that the effects due to the combination of agents may be detrimental or helpful, depending on the cell line studied. The data presented in this report complements the findings by Kauder et al. [29] and Blaskova et al. [44] regarding the effects of Aza-CdR in activating latently infected cells. In those studies, cell lines H12 and 2D12 [44] and all the J-Lat clones except for A2 clone [29] showed increased activation with Aza-CdR used in combination with TNFα in comparison with TNFα alone (the A2 clone showed the same levels of activation with or without Aza-CdR). The detrimental effect we observed with Aza-CdR in certain latently infected cell lines induced by viral activators like TNFα (ACH-2, J1.1 and U1) highlights the complexity of HIV reactivation, the importance of studies utilizing a broad range of cell lines, and the broad range of effects that may be observed in different infected cells.

Figure 4
figure 4

HIV activation by TNFα and TSA in ACH-2, U1, and J1.1 latently infected cell lines with or without Aza-CdR. A. p24 antigen production in ACH-2, U1 and J1.1 cell lines for all treatments and treatment combinations at 48 h post-induction. Cells were cultured in RPMI with 10% FBS. 2 million cells were treated with the different compounds for 48 h. The compound order of addition was determined in Fig. 1. After 48 h, cells were pelleted at 1400 rpm for 7 min and discarded. Supernatants from the different treatment conditions were stored at -80°C until further use. p24 antigen was determined using ELISA (Perkin Elmer, Waltham, MA) from the stored supernatants. B. Cell viability of the different treatments and treatment combinations shown in panel A, determined with an MTS assay (Promega, Madison, WI) at 48 h post-treatment. Compound final concentrations of the activators were Aza-CdR (2.5 μM), TNFα (10 ng/ml) and TSA (1.5 μM). Results are the mean ± standard deviation (SD). Statistical analysis (Student's t-test) was performed using STATA software package (StataCorp LP, College Station, TX), * p ≤ 0.05.

Table 1 Effect of AZA-CdR in TNFα HIV latently infected cell activation

The differential effects on HIV activation of epigenetic agents may not be entirely unexpected, since the chromatin structure of the LTR can differ for the proviruses integrated into one cell line or another [53]. It will require much additional work to understand the mechanisms responsible for the differential responses to epigenetic activators, but a few potential explanations may be considered. Aza-CdR may have complicated effects, since the activation of some genes may inhibit others [54–56], so differences in the methylation and hence activation state of cellular genes in the latently infected cell lines could account for the differences in activation observed with Aza-CdR: In J-Lat 6.3, 8.4 and 9.2 cells, Aza-CdR may have direct effects, leading to the demethylation of the LTR, enhancing activation, while in the other lines Aza-CdR may help activate other cellular genes that directly or indirectly inhibit HIV activation, since differential expression of certain cellular genes can be associated with the maintenance of latency [57, 58].

In HIV infected patients, the latently infected cells harbor many different proviruses. While the frequency of latently infected cells in the periphery is low, perhaps 1 in 106 cells, the total number of latently infected cells within a patient has been estimated to be as high as 106-107 total cells [59]. The latently infected lines we examined may not be completely representative of latently infected cells as they exist in vivo, but the great potential variability in host cell chromosomal location and epigenetic and transcriptional environments of proviruses from as many as 106-107 total latently infected cells suggests that, at least for some latently infected cells that exist in vivo, DNMTIs and HDACIs may inhibit HIV activation, as we describe here. While it is clear that DNMTIs and HDACIs offer promise as agents to attack the reservoir, our findings suggest that it may be necessary to carefully optimize HIV activation strategies so that some treatment or combination of treatments is active across large numbers of latently infected cells. For effective clinical applications, fairly elaborate combinations of activators and co-activators may be required to assure that essentially all proviruses are induced into active replication.

Abbreviations

Aza-CdR:

5-aza-2'deoxycytidine

DNMT:

DNA methyltransferase DNMTI: DNMT inhibitor

GFP:

green fluorescent protein

HAART:

highly active antiretroviral therapy

HDAC:

histone deacetylase

HDACI:

HDAC inhibitor

HIV-1:

Human Immunodeficiency Virus type I

LTR:

Long Terminal Repeat

MTS:

3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt

SAHA:

suberoylanilide hydroxamic acid

TAR:

Trans-activation response element

TNFα:

tumor necrosis factor alpha

TPA:

12-O-tetradecanoylphorbol-13-acetate

TSA:

trichostatin A

References

  1. Bukrinsky M, Stanwick T, Dempsey M, Stevenson M: Quiescent T lymphocytes as an inducible virus reservoir in HIV-1 infection. Science 1991, 254: 423-427. 10.1126/science.1925601

    Article  PubMed  CAS  Google Scholar 

  2. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, Lloyd AL, Nowak MA, Fauci AS: Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci USA 1997, 94: 13193-13197. 10.1073/pnas.94.24.13193

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  3. 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. 10.1126/science.278.5341.1295

    Article  PubMed  CAS  Google Scholar 

  4. Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD: Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997, 278: 1291-1295. 10.1126/science.278.5341.1291

    Article  PubMed  CAS  Google Scholar 

  5. Dinoso JB, Kim SY, Wiegand AM, Palmer SE, Gange SJ, Cranmer L, O'Shea A, Callender M, Spivak A, Brennan T, et al.: Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci USA 2009, 106: 9403-9408. 10.1073/pnas.0903107106

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  6. Colin L, Van Lint C: Molecular control of HIV-1 postintegration latency: implications for the development of new therapeutic strategies. Retrovirology 2009, 6: 111. 10.1186/1742-4690-6-111

    Article  PubMed  PubMed Central  Google Scholar 

  7. 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. 10.1126/science.1165706

    Article  PubMed  CAS  Google Scholar 

  8. Dahl V, Josefsson L, Palmer S: HIV reservoirs, latency, and reactivation: prospects for eradication. Antiviral Res 2010, 85: 286-294. 10.1016/j.antiviral.2009.09.016

    Article  PubMed  CAS  Google Scholar 

  9. Coiras M, Lopez-Huertas MR, Perez-Olmeda M, Alcami J: Understanding HIV-1 latency provides clues for the eradication of long-term reservoirs. Nat Rev Microbiol 2009, 7: 798-812. 10.1038/nrmicro2223

    Article  PubMed  CAS  Google Scholar 

  10. Marsden MD, Zack JA: Eradication of HIV: current challenges and new directions. J Antimicrob Chemother 2009, 63: 7-10. 10.1093/jac/dkn455

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  11. Geeraert L, Kraus G, Pomerantz RJ: Hide-and-seek: the challenge of viral persistence in HIV-1 infection. Annu Rev Med 2008, 59: 487-501. 10.1146/annurev.med.59.062806.123001

    Article  PubMed  CAS  Google Scholar 

  12. Hamer DH: Can HIV be Cured? Mechanisms of HIV persistence and strategies to combat it. Curr HIV Res 2004, 2: 99-111. 10.2174/1570162043484915

    Article  PubMed  CAS  Google Scholar 

  13. 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. 10.1182/blood.V98.10.3006

    Article  PubMed  CAS  Google Scholar 

  14. Dybul M, Hidalgo B, Chun TW, Belson M, Migueles SA, Justement JS, Herpin B, Perry C, Hallahan CW, Davey RT, et al.: Pilot study of the effects of intermittent interleukin-2 on human immunodeficiency virus (HIV)-specific immune responses in patients treated during recently acquired HIV infection. J Infect Dis 2002, 185: 61-68. 10.1086/338123

    Article  PubMed  CAS  Google Scholar 

  15. Chun TW, Engel D, Mizell SB, Hallahan CW, Fischette M, Park S, Davey RT Jr, Dybul M, Kovacs JA, Metcalf JA, et al.: Effect of interleukin-2 on the pool of latently infected, resting CD4+ T cells in HIV-1-infected patients receiving highly active anti-retroviral therapy. Nat Med 1999, 5: 651-655. 10.1038/9498

    Article  PubMed  CAS  Google Scholar 

  16. Stellbrink HJ, van Lunzen J, Westby M, O'Sullivan E, Schneider C, Adam A, Weitner L, Kuhlmann B, Hoffmann C, Fenske S, et al.: Effects of interleukin-2 plus highly active antiretroviral therapy on HIV-1 replication and proviral DNA (COSMIC trial). AIDS 2002, 16: 1479-1487. 10.1097/00002030-200207260-00004

    Article  PubMed  CAS  Google Scholar 

  17. Wang FX, Xu Y, Sullivan J, Souder E, Argyris EG, Acheampong EA, Fisher J, Sierra M, Thomson MM, Najera R, et al.: 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 2005, 115: 128-137.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  18. Lehrman G, Ylisastigui L, Bosch RJ, Margolis DM: Interleukin-7 induces HIV type 1 outgrowth from peripheral resting CD4+ T cells. J Acquir Immune Defic Syndr 2004, 36: 1103-1104. 10.1097/00126334-200408150-00015

    Article  PubMed  Google Scholar 

  19. Chun TW, Engel D, Mizell SB, Ehler LA, Fauci AS: Induction of HIV-1 replication in latently infected CD4+ T cells using a combination of cytokines. J Exp Med 1998, 188: 83-91. 10.1084/jem.188.1.83

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  20. Lafeuillade A, Poggi C, Chadapaud S, Hittinger G, Chouraqui M, Pisapia M, Delbeke E: Pilot study of a combination of highly active antiretroviral therapy and cytokines to induce HIV-1 remission. J Acquir Immune Defic Syndr 2001, 26: 44-55.

    Article  PubMed  CAS  Google Scholar 

  21. Chun TW, Davey RT Jr, Engel D, Lane HC, Fauci AS: Re-emergence of HIV after stopping therapy. Nature 1999, 401: 874-875. 10.1038/44755

    Article  PubMed  CAS  Google Scholar 

  22. Van Lint C, Emiliani S, Ott M, Verdin E: Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. Embo J 1996, 15: 1112-1120.

    PubMed  CAS  PubMed Central  Google Scholar 

  23. Sheridan PL, Mayall TP, Verdin E, Jones KA: Histone acetyltransferases regulate HIV-1 enhancer activity in vitro. Genes Dev 1997, 11: 3327-3340. 10.1101/gad.11.24.3327

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  24. 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. 10.1089/aid.2008.0191

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  25. Bohan C, York D, Srinivasan A: Sodium butyrate activates human immunodeficiency virus long terminal repeat--directed expression. Biochem Biophys Res Commun 1987, 148: 899-905. 10.1016/S0006-291X(87)80217-6

    Article  PubMed  CAS  Google Scholar 

  26. Laughlin M, Zeichner S, Kolson D, Alwine J, Seshamma T, Pomerantz R, Gonzalez-Scaran F: Sodium butryate treatment of cells latently infected with HIV-1 results in the expression of unspliced viral RNA. Virology 1993, 196: 496-505. 10.1006/viro.1993.1505

    Article  PubMed  CAS  Google Scholar 

  27. Moog C, Kuntz-Simon G, Caussin-Schwemling C, Obert G: Sodium valproate, an anticonvulsant drug, stimulates human immunodeficiency virus type 1 replication independently of glutathione levels. J Gen Virol 1996,77(Pt 9):1993-1999. 10.1099/0022-1317-77-9-1993

    Article  PubMed  CAS  Google Scholar 

  28. Archin NM, Eron JJ, Palmer S, Hartmann-Duff A, Martinson JA, Wiegand A, Bandarenko N, Schmitz JL, Bosch RJ, Landay AL, et al.: Valproic acid without intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4+ T cells. AIDS 2008, 22: 1131-1135. 10.1097/QAD.0b013e3282fd6df4

    Article  PubMed  CAS  Google Scholar 

  29. Kauder SE, Bosque A, Lindqvist A, Planelles V, Verdin E: Epigenetic regulation of HIV-1 latency by cytosine methylation. PLoS Pathog 2009, 5: e1000495. 10.1371/journal.ppat.1000495

    Article  PubMed  PubMed Central  Google Scholar 

  30. Quivy V, Adam E, Collette Y, Demonte D, Chariot A, Vanhulle C, Berkhout B, Castellano R, de Launoit Y, Burny A, et al.: Synergistic activation of human immunodeficiency virus type 1 promoter activity by NF-kappaB and inhibitors of deacetylases: potential perspectives for the development of therapeutic strategies. J Virol 2002, 76: 11091-11103. 10.1128/JVI.76.21.11091-11103.2002

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  31. Yedavalli VR, Jeang KT: Methylation: a regulator of HIV-1 replication? Retrovirology 2007, 4: 9. 10.1186/1742-4690-4-9

    Article  PubMed  PubMed Central  Google Scholar 

  32. Savarino A, Mai A, Norelli S, El Daker S, Valente S, Rotili D, Altucci L, Palamara AT, Garaci E: "Shock and kill" effects of class I-selective histone deacetylase inhibitors in combination with the glutathione synthesis inhibitor buthionine sulfoximine in cell line models for HIV-1 quiescence. Retrovirology 2009, 6: 52. 10.1186/1742-4690-6-52

    Article  PubMed  PubMed Central  Google Scholar 

  33. Verdin E, Paras P Jr, Van Lint C: Chromatin disruption in the promoter of human immunodeficiency virus type 1 during transcriptional activation. Embo J 1993, 12: 3249-3259.

    PubMed  CAS  PubMed Central  Google Scholar 

  34. El Kharroubi A, Piras G, Zensen R, Martin MA: Transcriptional activation of the integrated chromatin-associated human immunodeficiency virus type 1 promoter. Mol Cell Biol 1998, 18: 2535-2544.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  35. Benkirane M, Chun RF, Xiao H, Ogryzko VV, Howard BH, Nakatani Y, Jeang KT: Activation of integrated provirus requires histone acetyltransferase. p300 and P/CAF are coactivators for HIV-1 Tat. J Biol Chem 1998, 273: 24898-24905. 10.1074/jbc.273.38.24898

    Article  PubMed  CAS  Google Scholar 

  36. Marban C, Suzanne S, Dequiedt F, de Walque S, Redel L, Van Lint C, Aunis D, Rohr O: Recruitment of chromatin-modifying enzymes by CTIP2 promotes HIV-1 transcriptional silencing. EMBO J 2007, 26: 412-423. 10.1038/sj.emboj.7601516

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  37. Jiang G, Espeseth A, Hazuda DJ, Margolis DM: c-Myc and Sp1 contribute to proviral latency by recruiting histone deacetylase 1 to the human immunodeficiency virus type 1 promoter. J Virol 2007, 81: 10914-10923. 10.1128/JVI.01208-07

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  38. Coull JJ, He G, Melander C, Rucker VC, Dervan PB, Margolis DM: Targeted derepression of the human immunodeficiency virus type 1 long terminal repeat by pyrrole-imidazole polyamides. J Virol 2002, 76: 12349-12354. 10.1128/JVI.76.23.12349-12354.2002

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  39. Lusic M, Marcello A, Cereseto A, Giacca M: Regulation of HIV-1 gene expression by histone acetylation and factor recruitment at the LTR promoter. Embo J 2003, 22: 6550-6561. 10.1093/emboj/cdg631

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  40. Hejnar J, Hajkova P, Plachy J, Elleder D, Stepanets V, Svoboda J: CpG island protects Rous sarcoma virus-derived vectors integrated into nonpermissive cells from DNA methylation and transcriptional suppression. Proc Natl Acad Sci USA 2001, 98: 565-569. 10.1073/pnas.98.2.565

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  41. Taniguchi Y, Nosaka K, Yasunaga J, Maeda M, Mueller N, Okayama A, Matsuoka M: Silencing of human T-cell leukemia virus type I gene transcription by epigenetic mechanisms. Retrovirology 2005, 2: 64. 10.1186/1742-4690-2-64

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lorincz MC, Schubeler D, Groudine M: Methylation-mediated proviral silencing is associated with MeCP2 recruitment and localized histone H3 deacetylation. Mol Cell Biol 2001, 21: 7913-7922. 10.1128/MCB.21.23.7913-7922.2001

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  43. Curradi M, Izzo A, Badaracco G, Landsberger N: Molecular mechanisms of gene silencing mediated by DNA methylation. Mol Cell Biol 2002, 22: 3157-3173. 10.1128/MCB.22.9.3157-3173.2002

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  44. 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. 10.1371/journal.ppat.1000554

    Article  PubMed  PubMed Central  Google Scholar 

  45. Jeeninga RE, Westerhout EM, van Gerven ML, Berkhout B: HIV-1 latency in actively dividing human T cell lines. Retrovirology 2008, 5: 37. 10.1186/1742-4690-5-37

    Article  PubMed  PubMed Central  Google Scholar 

  46. 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. 10.1093/emboj/cdg188

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  47. Perez VL, Rowe T, Justement JS, Butera ST, June CH, Folks TM: An HIV-1-infected T cell clone defective in IL-2 production and Ca2+ mobilization after CD3 stimulation. J Immunol 1991, 147: 3145-3148.

    PubMed  CAS  Google Scholar 

  48. Clouse KA, Powell D, Washington I, Poli G, Strebel K, Farrar W, Barstad P, Kovacs J, Fauci AS, Folks TM: Monokine regulation of human immunodeficiency virus-1 expression in a chronically infected human T cell clone. J Immunol 1989, 142: 431-438.

    PubMed  CAS  Google Scholar 

  49. Folks TM, Justement J, Kinter A, Dinarello CA, Fauci AS: Cytokine-induced expression of HIV-1 in a chronically infected promonocyte cell line. Science 1987, 238: 800-802. 10.1126/science.3313729

    Article  PubMed  CAS  Google Scholar 

  50. Emiliani S, Van Lint C, Fischle W, Paras P Jr, Ott M, Brady J, Verdin E: A point mutation in the HIV-1 Tat responsive element is associated with postintegration latency. Proc Natl Acad Sci USA 1996, 93: 6377-6381. 10.1073/pnas.93.13.6377

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  51. Emiliani S, Fischle W, Ott M, Van Lint C, Amella CA, Verdin E: Mutations in the tat gene are responsible for human immunodeficiency virus type 1 postintegration latency in the U1 cell line. J Virol 1998, 72: 1666-1670.

    PubMed  CAS  PubMed Central  Google Scholar 

  52. Poli G, Kinter A, Justement JS, Kehrl JH, Bressler P, Stanley S, Fauci AS: Tumor necrosis factor alpha functions in an autocrine manner in the induction of human immunodeficiency virus expression. Proc Natl Acad Sci USA 1990, 87: 782-785. 10.1073/pnas.87.2.782

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  53. Verdin E: DNase I-hypersensitive sites are associated with both long terminal repeats and with the intragenic enhancer of integrated human immunodeficiency virus type 1. J Virol 1991, 65: 6790-6799.

    PubMed  CAS  PubMed Central  Google Scholar 

  54. Karpf AR, Lasek AW, Ririe TO, Hanks AN, Grossman D, Jones DA: Limited gene activation in tumor and normal epithelial cells treated with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine. Mol Pharmacol 2004, 65: 18-27. 10.1124/mol.65.1.18

    Article  PubMed  CAS  Google Scholar 

  55. Yu JN, Xue CY, Wang XG, Lin F, Liu CY, Lu FZ, Liu HL: 5-AZA-2'-deoxycytidine (5-AZA-CdR) leads to down-regulation of Dnmt1o and gene expression in preimplantation mouse embryos. Zygote 2009, 17: 137-145. 10.1017/S0967199408005169

    Article  PubMed  CAS  Google Scholar 

  56. Bender CM, Pao MM, Jones PA: Inhibition of DNA methylation by 5-aza-2'-deoxycytidine suppresses the growth of human tumor cell lines. Cancer Res 1998, 58: 95-101.

    PubMed  CAS  Google Scholar 

  57. Krishnan V, Zeichner SL: Alterations in the expression of DEAD-box and other RNA binding proteins during HIV-1 replication. Retrovirology 2004, 1: 42. 10.1186/1742-4690-1-42

    Article  PubMed  PubMed Central  Google Scholar 

  58. Krishnan V, Zeichner SL: Host cell gene expression during human immunodeficiency virus type 1 latency and reactivation and effects of targeting genes that are differentially expressed in viral latency. J Virol 2004, 78: 9458-9473. 10.1128/JVI.78.17.9458-9473.2004

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  59. 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. 10.1038/387183a0

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This work was funded in part by a Children's Research Institute, Children's National Medical Center Research Advisory Committee award to SZ and by the District of Columbia Developmental Center for AIDS Research (DC CFAR). GF is a Beatriu de Pinos Fellow of the Departament d'Universitats, Recerca i Societat de la Informació de la Generalitat de Catalunya. The following reagents were obtained through the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH: J-Lat full length Clones 6.3, 8.4, 9.2 and 10.6 from Dr. Eric Verdin, ACH-2, J1.1 and U1/HIV-1 from Dr. Thomas Folks.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Steven L Zeichner.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

GF carried out the experiments, data analysis and drafted the manuscript. SZ participated in the design of the study and data analysis, and drafted the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Fernandez, G., Zeichner, S.L. Cell line-dependent variability in HIV activation employing DNMT inhibitors. Virol J 7, 266 (2010). https://doi.org/10.1186/1743-422X-7-266

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1743-422X-7-266

Keywords