Histone deacetylase inhibitors impair the elimination of HIV-infected cells by cytotoxic T-lymphocytes

Richard Brad Jones, Rachel O'Connor, Stefanie Mueller, Maria Foley, Gregory L Szeto, Dan Karel, Mathias Lichterfeld, Colin Kovacs, Mario A Ostrowski, Alicja Trocha, Darrell J Irvine, Bruce D Walker, Richard Brad Jones, Rachel O'Connor, Stefanie Mueller, Maria Foley, Gregory L Szeto, Dan Karel, Mathias Lichterfeld, Colin Kovacs, Mario A Ostrowski, Alicja Trocha, Darrell J Irvine, Bruce D Walker

Abstract

Resting memory CD4+ T-cells harboring latent HIV proviruses represent a critical barrier to viral eradication. Histone deacetylase inhibitors (HDACis), such as suberanilohydroxamic acid (SAHA), romidepsin, and panobinostat have been shown to induce HIV expression in these resting cells. Recently, it has been demonstrated that the low levels of viral gene expression induced by a candidate HDACi may be insufficient to cause the death of infected cells by viral cytopathic effects, necessitating their elimination by immune effectors, such as cytotoxic T-lymphocytes (CTL). Here, we study the impact of three HDACis in clinical development on T-cell effector functions. We report two modes of HDACi-induced functional impairment: i) the rapid suppression of cytokine production from viable T-cells induced by all three HDACis ii) the selective death of activated T-cells occurring at later time-points following transient exposures to romidepsin or, to a lesser extent, panobinostat. As a net result of these factors, HDACis impaired CTL-mediated IFN-γ production, as well as the elimination of HIV-infected or peptide-pulsed target cells, both in liquid culture and in collagen matrices. Romidepsin exerted greater inhibition of antiviral function than SAHA or panobinostat over the dose ranges tested. These data suggest that treatment with HDACis to mobilize the latent reservoir could have unintended negative impacts on the effector functions of CTL. This could influence the effectiveness of HDACi-based eradication strategies, by impairing elimination of infected cells, and is a critical consideration for trials where therapeutic interruptions are being contemplated, given the importance of CTL in containing rebound viremia.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Effects of HDACis on the…
Figure 1. Effects of HDACis on the viability of PBMC CD8+ and CD4+ T-cells.
A–C. PBMC were treated with HDACi drugs at the indicated concentrations for either 4 or 21 hours (drugs left in) then stained with Annexin-V-Fitc (stains phosphatidyl serine on apoptotic cells), 7-AAD (stains DNA of dead cells), CD4 pacific blue, and CD8 alexa-fluor 700 and analyzed by flow cytometry. A. Shown are representative flow cytometry data indicating gating on dead (Annexin-V+7-AAD+) and dying (Annexin-vbright7-AADdim/−) cells. B, C. Shown are summary data for treatments PBMC gated on CD8+ cells (left panel) or CD4+ cells (right panel) with romidepsin and panobinostat (B) or with SAHA (C) at the indicated doses for the indicated times. P values for comparisons between the multiple doses of romidepsin tested and the untreated condition were calculated by Kruskal-Wallis test and found to be significant for ex vivo CD4+ T-cells (p = 0.0080). Post-hoc Dunn's multiple comparison tests were performed for each of these and the indicated p values are adjusted for multiple testing. Other drug treatment conditions and cell types were not significant by Kruskal-Wallis tests. D. The effects of HDACi treatment on the viability of an HIV-Gag-SLYNTVATL-specific T-cell clone were determined in the same manner as for PBMC, including use of the same statistical tests. Error bars represent SD. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.
Figure 2. Romidepsin and panobinostat are disproportionately…
Figure 2. Romidepsin and panobinostat are disproportionately toxic to activated CD4+ and CD8+ T-cells.
A. HIV-Env-VL11 and HIV-Gag SL9 specific CD8+ CTL clones were isolated from subjects OM292 and OM9 respectively, and exposed to HDACis at the indicated concentrations for 4 hours. Cells were then washed to remove drugs and replated in fresh R10 medium supplemented with 50 U/ml IL-2 for an additional 17 hours. Cell viability was measured by 7-AAD staining and flow cytometry. B. PBMC from subject OM292 either directly ex vivo (left panel) or following 48 hours of stimulation with anti-CD3 and anti-CD28 (right panel) were exposed to HDACis at the indicated concentrations for 4 hours. Cells were then washed to remove drugs and replated in fresh R10 medium. Cell viability was then measured by 7-AAD staining and flow cytometry. For A and B, shown are graphs of mean values taken from triplicate wells ± SEM following subtraction of background (% 7-AAD+ in no drug controls). P values were calculated by two-way ANOVA with Dunnett's multiple comparison test (comparing to the no drug control) * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001. C. In parallel to the viability assays shown in A and B, ACH2 cells were treated with HDACis for 4 hours, then washed and replated in fresh medium. Shown are mean ± SEM values for % HIV-Gag+ cells as measured in triplicate by intracellular staining and flow cytometry.
Figure 3. HDAC inhibitors impair IFN-γ production…
Figure 3. HDAC inhibitors impair IFN-γ production from PMA/ionomycin stimulated CD4+ and CD8+ T-cells.
A, B. PBMC from subject OM292 were exposed to HDACis for 4 hours. Cells were then washed and cultured in fresh medium with PMA/ionomycin for an additional 5 hours in the presence of brefeldin A. A. IFN-γ production in CD4+ and CD8+ T-cells was measured by intracellular cytokine staining flow cytometry. Shown are mean ± SEM values. P values were calculated by two-way ANOVA with Dunnett's multiple comparison test (comparing to the no drug control) * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001. B. Viability in CD4+ and CD8+ T-cells was measured by Annexin-V and 7-AAD staining flow cytometry. Shown are mean ± SEM values.
Figure 4. HDAC inhibitors impair IFN-γ production…
Figure 4. HDAC inhibitors impair IFN-γ production from antigen-stimulated CD8+ T-cells.
A, B. PBMC or CTL clones from chronically HIV-infected ARV-treated subjects (OM265 and OM292) were cultured for 2 hours with the indicated drugs and then plated for IFN-γ ELISPOT assays with 12 hour peptide stimulation periods. A. Treated CTL clones were stimulated with overlapping 15mer peptides spanning the CMV-pp65 protein (left panel) or the MHC-I-A24 restricted HIV-Nef peptide ‘RW8’ (right panel). Each treatment condition was tested in duplicate. Shown are mean SFC/104 CTL clone cells for each condition with error bars representing SEM. B. Experiments analogous to those depicted in A were performed for 8 different optimal HIV CD8+ T-cell epitopes (5 for OM292 and 1 for OM265). Shown are summary data for all 8 responses depicting the mean SFC/106 PBMC (of triplicate wells) under different treatment conditions. For SAHA, panobinostat, and romidepsin conditions statistical significance for each drug was evaluated using two-way ANOVA tests, and p values were calculated using Dunnett's multiple comparison test to account for the use of three different drug concentrations. For IL-15SA p values were calculated using the Wilcoxon matched pair test. C. PBMC from subjects OM265 and OM292 were treated with SAHA or romidepsin at the indicated concentrations for 2 hours and then either: stimulated with peptides for 6 a hour ELISPOT assay, or washed, cultured for 24 hours in the absence of drugs, and stimulated with peptides for a 12 hours ELISPOT assay. P values were calculated by two-way ANOVA with Dunnett's multiple comparison test (comparing to the no drug control) * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.
Figure 5. HDAC inhibitors impair proliferation of…
Figure 5. HDAC inhibitors impair proliferation of virus-specific T-cells.
PBMC from 11 chronically HIV-infected subjects on suppressive ARV therapy were labeled with CFSE and treated with the indicated drugs for 4 hours. HIV-Gag or CMV-pp65 peptide pools were then added to final concentrations of 1 µg/ml/peptide. 48 hours later, cells were washed thoroughly, re-suspended in medium with 20 U/ml IL-2, and cultured for an additional 5 days. Cells were then stained with a viability dye and antibodies to CD3, CD8, and analyzed by flow cytometry. A. Shown are representative flow cytometry data of a CMV-pp65 response gated on viable, CD3+CD8+ lymphocytes depicting CFSE (diminution indicates proliferation) – x-axis by CD8 – y-axis. B–E. Summary data depicting frequencies of CFSEdim cells within viable, CD3+CD8+ or CD3+CD8− (CD4− T-cell) lymphocyte populations. F. Shown are data analogous to B–E, but from a separate experiment where peptides were added back with fresh medium following HDACi wash-out at 48 hours. P values were calculated by the Wilcoxon matched-pairs signed rank test.
Figure 6. HDAC inhibitors impair CTL killing…
Figure 6. HDAC inhibitors impair CTL killing of HIV-infected primary CD4+ cells.
CD4+ T-cells were enriched from HIV-uninfected A02+ donors and either infected with HIV JR-CSF or maintained as mock infected controls. 24 hours post-infection, these target cells were co-cultured with an HIV-Gag- or CMV-pp65-specific CTL clones at the indicated effector (clone)∶target (CD4+ cell) ratios for 16 hours. Cells were then stained with fluorochrome conjugated antibodies to CD8, CD4, and HIV-Gag (intracellular staining) and analyzed by flow cytometry. A. Shown is the gating strategy utilized for killing assays, with the placement of Gag+ and Gag+CD4dim gates determined based on a mock-infected control (right panel). B. Data from a representative experiment with indicated effector∶target ratios of an HIV-Gag-SLYNTVATL-specific T-cell clone. Panels C-F utilize total Gag+ cells and calculate a % Infected Cells Killed as (%Gag+ No Effectors - %Gag+ at Given E∶T ratio)/%Gag+ No Effectors*100, ex. in panel B %Infected Cells Killed at 1∶36 ratio = (22.1−6.7)/22.1*100 = 69.7%. C–F. Shown are the results from 4 independent experiments. In C CTL were treated with romidepsin or panobinostat at 50 nM or with SAHA at 500 nM for 2 hours, and then co-cultured with target cells without a washing step. In D–F CTL were treated with romidepsin or panobinostat at 25 nM or with SAHA at 500 nM for 6 hours, then washed thoroughly before initiating a 16 hour co-culture with target cells. D and E both utilize an HIV-Gag-SLYNTVATL-specific CTL clone at high and low E∶T ratios respectively. E utilizes an HIV-Gag-FLGKIWPSHK-specific CTL clone. C–F. Shown are means ± SEM of data from triplicate wells. P values were calculated by two-way ANOVA with Tukey's multiple comparison test.
Figure 7. Impairment of CTL killing is…
Figure 7. Impairment of CTL killing is sustained for at least 14 hours after removal of romidepsin or panobinostat.
An HIV-Gag-specific CTL clone was treated with panobinostat, romidepsin, or SAHA at the indicated doses for 5 hours, or maintained as an untreated control. Cells were then washed two times, resuspended in R10–50, and cultured for 4 or 14 hours. These CTL were then co-cultured with autologous HIV-infected CD4+ T-cells at a clone∶target ratio of 1∶10 for 8 hours, and levels of infection were measured as described above (see Fig. 4 legend). A. Shown are flow cytometry plots gated on the viable (FSC/SSC) CD8− population, and depicting HIV-Gag staining (x-axis) by CD4 staining (y-axis). For the plots show, CTL that had been treated with romidepsin were given a 14 hour wash-out period prior to co-culture with infected cells. B. Shown are summary flow cytometry for 4 hour wash-outs (left panels) and 14 hour wash-outs (right panels). Each condition was tested in duplicate. Error bars represent SEM. C. Portions of the CTL used in the above elimination assays were maintained in culture. At the conclusions of the 14 hour wash-out elimination assay, 29 hours in total since addition of drugs, the frequencies of dead (7-AAD+) and apoptotic (Annexin-V+) CTL were measured by flow cytometry. Shown are % Annexin-V+ following treatment with the indicated doses of drugs. Error bars represent SEM. P values were calculated by two-way ANOVA with Dunnett's multiple comparison test (comparing to the no drug control) * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.
Figure 8. Time-lapse microscopy of CTL killing…
Figure 8. Time-lapse microscopy of CTL killing of peptide pulsed BLCL target cells in 3D collagen matrices.
An HIV-Gag-SLYNTVATL-specific CTL clone was labeled with Alexa-Fluor555 conjugated cholera toxin subunit B either cultured with 500 nM SAHA or 25 nM romidepsin for 20 hours, or maintained as an untreated control. These effector cells were combined with SLYNTVATL peptide pulsed target cells, matched on the restricting allele, in a collagen matrix medium containing sytox green viability dye. These mixtures were then plated in three separate wells of an 8-well cover slip and imaged by time-lapse brightfield and fluorescent microscopy. A. Shown are representative fields of view from the no treatment (upper panel), 500 nM SAHA (middle panel), and 25 nM romidepsin (lower panel) conditions advancing in time from left to right. Time stamps are given in hh∶mm format. Clones described in the results are indicated with yellow arrows and killed target cells are indicated with white arrows in the upper right panel. B. The number of killed (sytox green positive) BLCL at T = 20 minutes were counted in each of the four fields of view acquired for each condition. Each field of view is plotted as a single point on the graph along with means and SD. P values were calculated by the Kruskal-Wallis test. The reported values are corrected for multiple testing using Dunn's multiple comparison test.

References

    1. Deeks SG (2011) HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med 62: 141–155.
    1. Deeks SG, Lewin SR, Havlir DV (2013) The end of AIDS: HIV infection as a chronic disease. Lancet 382: 1525–1533.
    1. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, et al. (1997) Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A 94: 13193–13197.
    1. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, et al. (1997) Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278: 1295–1300.
    1. Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, et al. (1997) Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278: 1291–1295.
    1. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, et al. (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.
    1. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, et al. (2009) HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 15: 893–900.
    1. Buzon MJ, Massanella M, Llibre JM, Esteve A, Dahl V, et al. (2010) HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 16: 460–465.
    1. Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, et al. (2012) Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487: 482–485.
    1. Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, et al. (2009) Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Hum Retroviruses 25: 207–212.
    1. Rasmussen TA, Schmeltz Sogaard O, Brinkmann C, Wightman F, Lewin SR, et al. (2013) Comparison of HDAC inhibitors in clinical development: effect on HIV production in latently infected cells and T-cell activation. Hum Vaccin Immunother 9: 993–1001.
    1. Wightman F, Ellenberg P, Churchill M, Lewin SR (2012) HDAC inhibitors in HIV. Immunol Cell Biol 90: 47–54.
    1. Shan L, Deng K, Shroff NS, Durand CM, Rabi SA, et al. (2012) Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity 36: 491–501.
    1. Licciardi PV, Karagiannis TC (2012) Regulation of immune responses by histone deacetylase inhibitors. ISRN Hematol 2012: 690901.
    1. Akimova T, Beier UH, Liu Y, Wang L, Hancock WW (2012) Histone/protein deacetylases and T-cell immune responses. Blood 119: 2443–2451.
    1. Bosisio D, Vulcano M, Del Prete A, Sironi M, Salvi V, et al. (2008) Blocking TH17-polarizing cytokines by histone deacetylase inhibitors in vitro and in vivo. J Leukoc Biol 84: 1540–1548.
    1. Glauben R, Batra A, Fedke I, Zeitz M, Lehr HA, et al. (2006) Histone hyperacetylation is associated with amelioration of experimental colitis in mice. J Immunol 176: 5015–5022.
    1. Leoni F, Zaliani A, Bertolini G, Porro G, Pagani P, et al. (2002) The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits antiinflammatory properties via suppression of cytokines. Proc Natl Acad Sci U S A 99: 2995–3000.
    1. Mishra N, Reilly CM, Brown DR, Ruiz P, Gilkeson GS (2003) Histone deacetylase inhibitors modulate renal disease in the MRL-lpr/lpr mouse. J Clin Invest 111: 539–552.
    1. Reddy P, Maeda Y, Hotary K, Liu C, Reznikov LL, et al. (2004) Histone deacetylase inhibitor suberoylanilide hydroxamic acid reduces acute graft-versus-host disease and preserves graft-versus-leukemia effect. Proc Natl Acad Sci U S A 101: 3921–3926.
    1. Shi ZJ, Ouyang DY, Zhu JS, Xu LH, He XH (2012) Histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits anti-inflammatory activities through induction of mitochondrial damage and apoptosis in activated lymphocytes. Int Immunopharmacol 12: 580–587.
    1. Brogdon JL, Xu Y, Szabo SJ, An S, Buxton F, et al. (2007) Histone deacetylase activities are required for innate immune cell control of Th1 but not Th2 effector cell function. Blood 109: 1123–1130.
    1. Ge Z, Da Y, Xue Z, Zhang K, Zhuang H, et al. (2013) Vorinostat, a histone deacetylase inhibitor, suppresses dendritic cell function and ameliorates experimental autoimmune encephalomyelitis. Exp Neurol 241: 56–66.
    1. Kelly-Sell MJ, Kim YH, Straus S, Benoit B, Harrison C, et al. (2012) The histone deacetylase inhibitor, romidepsin, suppresses cellular immune functions of cutaneous T-cell lymphoma patients. Am J Hematol 87: 354–360.
    1. Moreira JM, Scheipers P, Sorensen P (2003) The histone deacetylase inhibitor Trichostatin A modulates CD4+ T cell responses. BMC Cancer 3: 30.
    1. Wang B, Morinobu A, Horiuchi M, Liu J, Kumagai S (2008) Butyrate inhibits functional differentiation of human monocyte-derived dendritic cells. Cell Immunol 253: 54–58.
    1. Bohmig GA, Krieger PM, Saemann MD, Wenhardt C, Pohanka E, et al. (1997) n-butyrate downregulates the stimulatory function of peripheral blood-derived antigen-presenting cells: a potential mechanism for modulating T-cell responses by short-chain fatty acids. Immunology 92: 234–243.
    1. Camelo S, Iglesias AH, Hwang D, Due B, Ryu H, et al. (2005) Transcriptional therapy with the histone deacetylase inhibitor trichostatin A ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol 164: 10–21.
    1. Dowdell KC, Pesnicak L, Hoffmann V, Steadman K, Remaley AT, et al. (2009) Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, diminishes lymphoproliferation in the Fas -deficient MRL/lpr(−/−) murine model of autoimmune lymphoproliferative syndrome (ALPS). Exp Hematol 37: 487–494.
    1. Garcia BA, Busby SA, Shabanowitz J, Hunt DF, Mishra N (2005) Resetting the epigenetic histone code in the MRL-lpr/lpr mouse model of lupus by histone deacetylase inhibition. J Proteome Res 4: 2032–2042.
    1. Larsen L, Tonnesen M, Ronn SG, Storling J, Jorgensen S, et al. (2007) Inhibition of histone deacetylases prevents cytokine-induced toxicity in beta cells. Diabetologia 50: 779–789.
    1. Reddy P, Sun Y, Toubai T, Duran-Struuck R, Clouthier SG, et al. (2008) Histone deacetylase inhibition modulates indoleamine 2,3-dioxygenase-dependent DC functions and regulates experimental graft-versus-host disease in mice. J Clin Invest 118: 2562–2573.
    1. Wang D, Iclozan C, Liu C, Xia C, Anasetti C, et al. (2012) LBH589 enhances T cell activation in vivo and accelerates graft-versus-host disease in mice. Biol Blood Marrow Transplant 18: 1182–1190 e1181.
    1. Bridle BW, Chen L, Lemay CG, Diallo JS, Pol J, et al. (2013) HDAC inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity during tumor immunotherapy. Mol Ther 21: 887–894.
    1. Khan N, Jeffers M, Kumar S, Hackett C, Boldog F, et al. (2008) Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. Biochem J 409: 581–589.
    1. Liu T, Kuljaca S, Tee A, Marshall GM (2006) Histone deacetylase inhibitors: multifunctional anticancer agents. Cancer Treat Rev 32: 157–165.
    1. Marks PA, Richon VM, Miller T, Kelly WK (2004) Histone deacetylase inhibitors. Adv Cancer Res 91: 137–168.
    1. Lindemann RK, Gabrielli B, Johnstone RW (2004) Histone-deacetylase inhibitors for the treatment of cancer. Cell Cycle 3: 779–788.
    1. Mayo MW, Denlinger CE, Broad RM, Yeung F, Reilly ET, et al. (2003) Ineffectiveness of histone deacetylase inhibitors to induce apoptosis involves the transcriptional activation of NF-kappa B through the Akt pathway. J Biol Chem 278: 18980–18989.
    1. Johnstone RW (2002) Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov 1: 287–299.
    1. Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, et al. (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 13: 1299–1307.
    1. Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, et al. (1995) Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science 270: 1811–1815.
    1. Beier UH, Akimova T, Liu Y, Wang L, Hancock WW (2011) Histone/protein deacetylases control Foxp3 expression and the heat shock response of T-regulatory cells. Curr Opin Immunol 23: 670–678.
    1. Clouse KA, Powell D, Washington I, Poli G, Strebel K, et al. (1989) Monokine regulation of human immunodeficiency virus-1 expression in a chronically infected human T cell clone. J Immunol 142: 431–438.
    1. Foley MH, Forcier T, McAndrew E, Gonzalez M, Chen H, et al. (2014) High avidity CD8+ T cells efficiently eliminate motile hiv-infected targets and execute a locally focused program of anti-viral function. PLoS One 9: e87873.
    1. Rothstein TL, Mage M, Jones G, McHugh LL (1978) Cytotoxic T lymphocyte sequential killing of immobilized allogeneic tumor target cells measured by time-lapse microcinematography. J Immunol 121: 1652–1656.
    1. Sanderson CJ (1976) The mechanism of T cell mediated cytotoxicity. II. Morphological studies of cell death by time-lapse microcinematography. Proc R Soc Lond B Biol Sci 192: 241–255.
    1. Matter A (1979) Microcinematographic and electron microscopic analysis of target cell lysis induced by cytotoxic T lymphocytes. Immunology 36: 179–190.
    1. Corporation C (2013) Istodax Package Insert.
    1. Zhu X, Marcus WD, Xu W, Lee HI, Han K, et al. (2009) Novel human interleukin-15 agonists. J Immunol 183: 3598–3607.
    1. Han KP, Zhu X, Liu B, Jeng E, Kong L, et al. (2011) IL-15:IL-15 receptor alpha superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization. Cytokine 56: 804–810.
    1. Jennes W, Kestens L, Nixon DF, Shacklett BL (2002) Enhanced ELISPOT detection of antigen-specific T cell responses from cryopreserved specimens with addition of both IL-7 and IL-15–the Amplispot assay. J Immunol Methods 270: 99–108.
    1. Strengell M, Matikainen S, Siren J, Lehtonen A, Foster D, et al. (2003) IL-21 in synergy with IL-15 or IL-18 enhances IFN-gamma production in human NK and T cells. J Immunol 170: 5464–5469.
    1. Shapiro GI, Frank R, Dandamudi UB, Hengelage T, Zhao L, et al. (2012) The effect of food on the bioavailability of panobinostat, an orally active pan-histone deacetylase inhibitor, in patients with advanced cancer. Cancer Chemother Pharmacol 69: 555–562.
    1. Wei DG, Chiang V, Fyne E, Balakrishnan M, Barnes T, et al. (2014) Histone Deacetylase Inhibitor Romidepsin Induces HIV Expression in CD4 T Cells from Patients on Suppressive Antiretroviral Therapy at Concentrations Achieved by Clinical Dosing. PLoS Pathog 10: e1004071.
    1. Rubin EH, Agrawal NG, Friedman EJ, Scott P, Mazina KE, et al. (2006) A study to determine the effects of food and multiple dosing on the pharmacokinetics of vorinostat given orally to patients with advanced cancer. Clin Cancer Res 12: 7039–7045.
    1. Mejia EJ, Loveridge ST, Stepan G, Tsai A, Jones GS, et al. (2014) Study of Marine Natural Products Including Resorcyclic Acid Lactones from Humicola fuscoatra That Reactivate Latent HIV-1 Expression in an in Vitro Model of Central Memory CD4+ T Cells. J Nat Prod 77: 618–24..
    1. Jones RB, Yue FY, Gu XX, Hunter DV, Mujib S, et al. (2009) Human immunodeficiency virus type 1 escapes from interleukin-2-producing CD4+ T-cell responses without high-frequency fixation of mutations. J Virol 83: 8722–8732.
    1. Streeck H, Frahm N, Walker BD (2009) The role of IFN-gamma Elispot assay in HIV vaccine research. Nat Protoc 4: 461–469.
    1. Walker BD, Flexner C, Birch-Limberger K, Fisher L, Paradis TJ, et al. (1989) Long-term culture and fine specificity of human cytotoxic T-lymphocyte clones reactive with human immunodeficiency virus type 1. Proc Natl Acad Sci U S A 86: 9514–9518.
    1. Jones RB, Garrison KE, Mujib S, Mihajlovic V, Aidarus N, et al. (2012) HERV-K-specific T cells eliminate diverse HIV-1/2 and SIV primary isolates. J Clin Invest 122: 4473–4489.
    1. Sacha JB, Watkins DI (2010) Synchronous infection of SIV and HIV in vitro for virology, immunology and vaccine-related studies. Nat Protoc 5: 239–246.

Source: PubMed

3
Suscribir