82
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      An In-Depth Comparison of Latency-Reversing Agent Combinations in Various In Vitro and Ex Vivo HIV-1 Latency Models Identified Bryostatin-1+JQ1 and Ingenol-B+JQ1 to Potently Reactivate Viral Gene Expression

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The persistence of latently infected cells in patients under combinatory antiretroviral therapy (cART) is a major hurdle to HIV-1 eradication. Strategies to purge these reservoirs are needed and activation of viral gene expression in latently infected cells is one promising strategy. Bromodomain and Extraterminal (BET) bromodomain inhibitors (BETi) are compounds able to reactivate latent proviruses in a positive transcription elongation factor b (P-TEFb)-dependent manner. In this study, we tested the reactivation potential of protein kinase C (PKC) agonists (prostratin, bryostatin-1 and ingenol-B), which are known to activate NF-κB signaling pathway as well as P-TEFb, used alone or in combination with P-TEFb-releasing agents (HMBA and BETi (JQ1, I-BET, I-BET151)). Using in vitro HIV-1 post-integration latency model cell lines of T-lymphoid and myeloid lineages, we demonstrated that PKC agonists and P-TEFb-releasing agents alone acted as potent latency-reversing agents (LRAs) and that their combinations led to synergistic activation of HIV-1 expression at the viral mRNA and protein levels. Mechanistically, combined treatments led to higher activations of P-TEFb and NF-κB than the corresponding individual drug treatments. Importantly, we observed in ex vivo cultures of CD8+-depleted PBMCs from 35 cART-treated HIV-1+ aviremic patients that the percentage of reactivated cultures following combinatory bryostatin-1+JQ1 treatment was identical to the percentage observed with anti-CD3+anti-CD28 antibodies positive control stimulation. Remarkably, in ex vivo cultures of resting CD4+ T cells isolated from 15 HIV-1+ cART-treated aviremic patients, the combinations bryostatin-1+JQ1 and ingenol-B+JQ1 released infectious viruses to levels similar to that obtained with the positive control stimulation. The potent effects of these two combination treatments were already detected 24 hours post-stimulation. These results constitute the first demonstration of LRA combinations exhibiting such a potent effect and represent a proof-of-concept for the co-administration of two different types of LRAs as a potential strategy to reduce the size of the latent HIV-1 reservoirs.

          Author Summary

          Persistence of latently infected cells during cART is a major hurdle for HIV-1 eradication. A widely proposed strategy to purge these reservoirs involves the reactivation of latent proviruses. The low levels of active P-TEFb and the cytoplasmic sequestration of NF-κB in resting infected cells largely contribute to maintenance of HIV-1 latency. Therefore, utilization of chemical compounds that target both pathways may lead to more potent effects on HIV-1 reactivation than the effect mediated by the individual drug treatments. In this study, we showed that combined treatments of PKC agonists (prostratin, bryostatin-1 and ing-B) with compounds releasing P-TEFb (JQ1, I-BET, I-BET151 and HMBA) exhibited a synergistic increase in viral reactivation from latency. In-depth comparison of combined treatments in various in vitro cellular models of HIV-1 latency as well as in ex vivo primary cell cultures from cART-treated HIV + aviremic patients identified bryostatin-1+JQ1 and ing-B+JQ1 to potently reactivate latent HIV-1. The potent effects of these two combinations were detected as early as 24 hours post-treatment. Importantly, bryostatin-1 was used at concentrations below the drug plasma levels achieved by doses used in children with refractory solid tumors. Our mechanistic data established a correlation between potentiated P-TEFb activation and potentiated or synergistic (depending on the HIV-1 latency cellular model used) induction of HIV-1 gene expression observed after the combined versus individual drug treatments. In conclusion, our results establish a proof-of-concept for PKC agonists combined with compounds releasing active P-TEFb as a strategy proposed for a cure or a durable remission of HIV infection.

          Related collections

          Most cited references76

          • Record: found
          • Abstract: found
          • Article: not found

          Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure.

          Antiretroviral therapy fails to cure HIV-1 infection because latent proviruses persist in resting CD4(+) T cells. T cell activation reverses latency, but <1% of proviruses are induced to release infectious virus after maximum in vitro activation. The noninduced proviruses are generally considered defective but have not been characterized. Analysis of 213 noninduced proviral clones from treated patients showed 88.3% with identifiable defects but 11.7% with intact genomes and normal long terminal repeat (LTR) function. Using direct sequencing and genome synthesis, we reconstructed full-length intact noninduced proviral clones and demonstrated growth kinetics comparable to reconstructed induced proviruses from the same patients. Noninduced proviruses have unmethylated promoters and are integrated into active transcription units. Thus, it cannot be excluded that they may become activated in vivo. The identification of replication-competent noninduced proviruses indicates that the size of the latent reservoir-and, hence, the barrier to cure-may be up to 60-fold greater than previously estimated. Copyright © 2013 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation.

            HIV persists in a reservoir of latently infected CD4(+) T cells in individuals treated with highly active antiretroviral therapy (HAART). Here we identify central memory (T(CM)) and transitional memory (T(TM)) CD4(+) T cells as the major cellular reservoirs for HIV and find that viral persistence is ensured by two different mechanisms. HIV primarily persists in T(CM) cells in subjects showing reconstitution of the CD4(+) compartment upon HAART. This reservoir is maintained through T cell survival and low-level antigen-driven proliferation and is slowly depleted with time. In contrast, proviral DNA is preferentially detected in T(TM) cells from aviremic individuals with low CD4(+) counts and higher amounts of interleukin-7-mediated homeostatic proliferation, a mechanism that ensures the persistence of these cells. Our results suggest that viral eradication might be achieved through the combined use of strategic interventions targeting viral replication and, as in cancer, drugs that interfere with the self renewal and persistence of proliferating memory T cells.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Novel ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo

              HIV-1 persists in a latent reservoir (LR) despite antiretroviral therapy (ART) 1–5 . This reservoir is the major barrier to HIV-1 eradication 6,7 . Current approaches to purging the LR involve pharmacologic induction of HIV-1 transcription and subsequent killing of infected cells by cytolytic T lymphocytes (CTL) or viral cytopathic effects 8–10 . Agents that reverse latency without activating T cells have been identified using in vitro models of latency. However, their effects on latently infected cells from infected individuals remain largely unknown. Using a novel ex vivo assay, we demonstrate that none of the latency reversing agents (LRAs) tested induced outgrowth of HIV-1 from the LR of patients on ART. Using a novel RT-qPCR assay specific for all HIV-1 mRNAs, we demonstrate that LRAs that do not cause T cell activation do not induce significant increases in intracellular HIV-1 mRNA in patient cells; only the PKC agonist bryostatin-1 caused substantial increases. These findings demonstrate that current in vitro models do not fully recapitulate mechanisms governing HIV-1 latency in vivo. Further, our data indicate that non-activating LRAs are unlikely to drive the elimination of the LR in vivo when administered individually. HIV-1 cure is hindered by viral persistence in a small fraction (~1/106) of resting CD4+ T cells (rCD4s) that harbor latent but replication-competent proviruses 1–3 . Upon cellular activation, latency is reversed and replication-competent virus is produced. Although T cell activation reverses latency, global T cell activation is toxic, generating interest in small molecule latency-reversing agents (LRAs) that do not activate T cells. Due to the low frequency of latently infected rCD4s in vivo, cell models have been used to identify a number of mechanistically distinct LRAs. These include: (1) histone deacetylase (HDAC) inhibitors, thought to function through epigenetic and other mechanisms 11–14 ; (2) disulfiram, postulated to involve nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) 15,16 ; and (3) the bromodomain-containing protein 4 (BRD4) inhibitor JQ1, which elicits effects through positive transcription elongation factor (P-TEFb) 17–20 . Acting through signaling pathways associated with T cell activation, protein kinase C (PKC) agonists such as phorbol esters, prostratin 21–23 and bryostatin-1 12,24–26 also reverse latency in cell models. Evidence that putative LRAs reverse latency ex vivo in primary rCD4s from HIV-1-infected individuals is limited; disulfiram and the HDAC inhibitor vorinostat have been tested in patient cells with inconsistent results 11,13,16,27,28 . Clinical trials in patients on ART are ongoing with disulfiram and the HDAC inhibitors vorinostat, romidepsin, and panobinostat 27,29 . A recent trial of disulfiram showed no consistent evidence of latency reversal 30 . In another clinical trial, a single dose of vorinostat modestly increased intracellular RNAs containing HIV-1 gag sequences in rCD4s of patients on ART 27 . Ex vivo treatment of patient cells with vorinostat induced outgrowth in some studies 11,13 but no virion production in another study 28 . Importantly, no LRA has been shown to reduce the size of the LR. A consistent ex vivo validation strategy has not been employed to compare putative LRAs. Given the costs and risks associated with clinical trials, such a strategy is important for HIV-1 eradication research. Therefore, we utilized three independent assays to evaluate the efficacy of LRAs in cells from HIV-1 infected individuals on suppressive ART (participant characteristics in Supplementary Table 1). We first tested LRAs in a modified viral outgrowth assay 1 . In the original assay, patient-derived rCD4s were activated and co-cultured with CD4+ T lymphoblasts from healthy donors to expand released virus. Induction of outgrowth provides conclusive evidence of latency reversal. In the modified assay, T cell activation was replaced with LRA treatment. The subsequent co-culture of patient rCD4s with healthy donor lymphoblasts constitutes a mixed lymphocyte reaction, which induces background reactivation of latent HIV-1 31 and complicates LRA evaluation. Therefore, we treated rCD4s with LRAs and then cultured the cells with a transformed CD4+ T cell line (MOLT-4/CCR5) (Fig. 1a) that supports robust HIV-1 replication but does not induce allogeneic stimulation of rCD4s (Supplementary Fig. 1a–c). We treated five million purified rCD4s from infected individuals on ART with single LRAs for 18 h and then co-cultured the cells with MOLT-4/CCR5 cells for 14 days to permit viral outgrowth. T cell activation with phorbol 12-myristate 13-acetate + ionomycin (PMA/I) served as a positive control. We concurrently measured the frequency of latently infected cells 32 . We evaluated vorinostat, romidepsin, panobinostat, disulfiram and bryostatin-1 at clinically relevant concentrations that effectively reversed latency in a primary cell model (see below) and that were not toxic to rCD4s. No drug treatment induced cell death as shown by the lack of 7-AAD staining (Fig. 1b). Surprisingly, none of the LRAs induced viral outgrowth from cells from any individual tested while PMA/I-treated cultures were positive for every patient with a detectable LR (Fig. 1c). We next asked whether LRA treatment induced rapid virus release. We collected culture supernatants from rCD4s from five infected individuals (S26–S30) after 18 h of LRA treatment and prior to addition of MOLT-4/CCR5 cells for measurement of viral outgrowth. PMA/I induced virus release as detected by HIV-1 mRNA in the supernatant from four out of five individuals (S26–S29) (Fig. 1D). Bryostatin-1 treatment induced detectable supernatant HIV-1 mRNA from one infected individual (S27), whereas no other LRA had a measurable effect (Fig. 1d). None of the LRAs induced subsequent viral outgrowth from these treated cells, including the cells from the single individual (S27) that released HIV-1 mRNA after bryostatin-1 treatment (Fig. 1c). The most widely used method to detect induction of HIV-1 transcription 16,27 in cells from infected individuals involves the measurement of RNAs containing HIV-1 gag sequences. Because this method lacks a stringent selection for poly-adenylated RNAs, it does not exclusively detect fully elongated and correctly processed HIV-1 mRNAs. Therefore, we devised a new assay specific for intracellular HIV-1 mRNA using a primer/probe set that detects the 3′ sequence common to all correctly terminated HIV-1 mRNAs (Fig. 2a). We detected baseline intracellular HIV-1 mRNA in rCD4s from ten out of 11 infected individuals. Stimulation with PMA/I for 18 h dramatically increased intracellular HIV-1 mRNA (mean increase = 115.5-fold, Fig. 2b). However, at clinically relevant concentrations that reverse latency in a primary cell model (Fig. 3B, C), vorinostat, romidepsin, panobinostat, disulfiram, and JQ1 failed to increase intracellular HIV-1 mRNA in rCD4s from infected individuals when used as single agents (Fig. 2b, c). Bryostatin- 1 caused significant increases in some infected individuals (Fig. 2c). We observed similar results after 6 h of LRA treatment (Supplementary Fig. 2). While no effect was seen in latently infected cells from infected individuals, LRA treatment increased intracellular HIV-1 mRNA in a B-cell lymphoma 2 (BCL-2) transduced primary rCD4 model of latency (Fig. 3a). LRA-induced increases in HIV-1 mRNA were consistent with measurements of the fraction of cells that up-regulate HIV-1 gene expression, as assessed by GFP reporter (Fig. 3b). The frequency of latent infection in this model is substantially higher than that observed in vivo 4 . To confirm that our assay effectively detects intracellular HIV-1 mRNA increases at frequencies of latent infection seen in vivo, we treated model cells with a known percentage of latent infection and then serially diluted these cells into rCD4s from uninfected individuals immediately prior to RNA isolation. We detected proportionate increases in intracellular HIV-1 mRNA in vorinostat-treated cells down to a frequency of 1/106 cells (Fig. 2d, e). Therefore, the lack of LRA efficacy in cells from HIV-1 infected individuals is not a result of assay insensitivity. Rather, our findings demonstrate that freshly isolated latently infected cells from infected individuals responded differently to LRAs than latency model cells. RT-qPCR assays that detect gag-containing sequences in total RNA are frequently used to detect latency reversal. These sequences do not necessarily represent bona fide unspliced HIV-1 mRNA. HIV-1 integrates into host genes that are actively transcribed in rCD4s 33,34 , allowing for the production of chimeric host/HIV-1 primary transcripts. Such transcripts, initiated at host promoters, could contain gag sequence and would be indistinguishable from LTR-initiated transcripts by conventional gag RT-qPCR assays (Fig. 4a). We therefore designed a primer/probe set that amplifies a region of the LTR that is not transcribed during LTR-initiated and correctly terminated HIV-1 transcription. This primer/probe set is specific for transcripts containing read-through of the 5′ LTR or 3′ LTR, independent of proviral orientation (Fig. 4a). We treated ten million rCD4s from infected individuals on ART with vorinostat or PMA/I for 6 h and compared the levels of HIV-1 mRNA, read-through transcripts, and transcripts containing gag sequence (Fig. 4a, b). We detected a small increase (~2-fold) in transcripts containing gag sequence in vorinostat-treated rCD4s from four out of five infected individuals, consistent with previous reports 27 (Fig. 4b). Vorinostat treatment also induced increases in read-through transcripts (Fig. 4b) comparable to the increases in transcripts containing gag sequence but had no effect on levels of HIV-1 mRNA (Fig. 4b). To prove that the read-through signal is amplified from a transcript that initiated upstream of the 5′ LTR and contains gag sequence, we primed cDNA synthesis with a gag primer (Fig. 4c). We detected comparable, statistically significant inductions of read-through and gag transcripts after 6 h of vorinostat treatment (Fig. 4d) (P = 0.027, P = 0.011, respectively; ratio paired t-test of transcript copies), indicative of read-through transcription. PMA/I induction of gag transcripts greatly exceeded that of read-through transcripts, indicative of LTR-initiated transcription (Supplementary Fig. 3). While not every potential LRA will induce read-through transcription by activating a host gene, our data show that chimeric host/HIV-1 transcripts can have a confounding effect on the RT-qPCR signal obtained with standard gag primers. Such an effect should be taken into consideration when evaluating LRAs using conventional gag RT-qPCR assays. The novel assays presented herein facilitated the first comparative ex vivo evaluation of candidate LRAs. Our data demonstrate that none of the leading candidate non-T cell activating LRAs tested significantly disrupted the LR ex vivo. The striking discordance between the effects of non-stimulating LRAs in in vitro models of HIV-1 latency and the ex vivo effects in rCD4s from infected individuals on ART indicates that these models do not fully capture all mechanisms governing HIV-1 latency in vivo. These compounds are unlikely to drive the elimination of the LR in vivo when administered individually. The only active single agent was the PKC agonist bryostatin-1, which is likely too toxic for clinical use. Whether other PKC agonists or other compounds that stimulate signaling pathways associated with T cell activation can be safely administered remains to be seen, and further progress may depend on finding safe and active combinations of LRAs. Methods Cell isolation and culture The Johns Hopkins Institutional Review Board approved this study and all research participants in this study gave written informed consent. Infected individuals were enrolled under the criteria of suppression of viremia to undetectable levels ( 0.5. We determined that the limit of quantification for all transcripts was 10 copies. A PCR signal of less than 10 copies (1–9 copies) was treated as 10 copies in calculations of fold change and marked as 10 copies on graphs depicting RNA copies. Undetectable PCR signal was treated as 10 copies in calculations of fold change and marked as 1 copy on graphs depicting RNA copies. Levels of RNA polymerase II (Pol2) and Glucose-6-phosphate dehydrogenase (G6PD) RNA were also measured for each sample to use as an endogenous control. Voronistat, romidepsin, panobinostat, JQ1 and PMA/I treatment consistently increased expression Pol2 and G6PD. Samples treated with the same drug had even levels of Pol2 and G6PD, indicating that the template inputs were approximately equal. Measurement of supernatant HIV-1 mRNA HIV-1 mRNA was extracted from 0.2mL of supernatant from five million cultured rCD4s after 18 h of LRA treatment using the ZR-96 Viral RNA kit (Zymo Research). cDNA synthesis was performed using qScript cDNA Supermix (Quanta Biosciences). Real-time PCR was performed using TaqMan Fast Advanced mastermix (Applied Biosystems) on an ABI Viia 7 Real-Time PCR machine. Primers and probes listed below. Manufacturer’s thermal cycling conditions were used. Molecular standard curve was generated as described above. Primer and probe sequences Nucleotide coordinates are indicated relative to HXB2 consensus sequence. HIV-1 mRNAs were detected using the following primers and probe, modified from Shan et al. 37 : Forward (5′→3′) CAGATGCTGCATATAAGCAGCTG (9501–9523) Reverse (5′→3′) TTTTTTTTTTTTTTTTTTTTTTTTGAAGCAC (9629-poly A) Probe (5′→3′) FAM-CCTGTACTGGGTCTCTCTGG-MGB (9531–9550) Transcripts containing HIV-1 gag sequence were detected using the following primers and probe, described previously 27 . Forward (5′→3′) ACATCAAGCAGCCATGCAAAT (1368–1388) Reverse (5′→3′) TCTGGCCTGGTGCAATAGG (1453–1471) Probe (5′→3′) VIC-CTATCCCATTCTGCAGCTTCCTCATTGATG-TAMRA (1401–1430) Chimeric host/HIV-1 read-through transcripts were detected using the following primers and probe: Forward (5′→3′) CAGATGCTGCATATAAGCAGCTG (416–438, 9501–9523) Reverse (5′→3′) CACAACAGACGGGCACACAC (556–575, 9641–9660) Probe (5′→3′) FAM-CCTGTACTGGGTCTCTCTGG-MGB (446–465, 9531–9550) cDNA synthesis reaction with gag primer sequence: Reverse (5′→3′) GTCACTTCCCCTTGG (1480–1494) Supplementary Material 1
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Pathog
                PLoS Pathog
                plos
                plospath
                PLoS Pathogens
                Public Library of Science (San Francisco, CA USA )
                1553-7366
                1553-7374
                30 July 2015
                July 2015
                : 11
                : 7
                : e1005063
                Affiliations
                [1 ]Service of Molecular Virology, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
                [2 ]Service des Maladies Infectieuses, Université de Liège, Centre Hospitalier Universitaire (CHU) de Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
                [3 ]Departments of Medicine, Microbiology, and Immunology, University of California, San Francisco, San Francisco, California, United States of America
                [4 ]Laboratory of Immunology, Brugmann University Hospital, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
                [5 ]Institut Universitaire de Technologie Louis Pasteur de Schiltigheim, University of Strasbourg, Schiltigheim, France
                [6 ]Service de Virologie, Université Paris-Descartes, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
                [7 ]Service des Maladies Infectieuses, CHU St-Pierre, ULB, Bruxelles, Belgium
                [8 ]Service de Génétique, Centre Hospitalier Universitaire (CHU) de Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
                [9 ]IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Illkirch-Graffenstaden, France
                [10 ]Kyolab, Rua Isaura Ap. Oliviera Barbosa Terini, Sao Paulo, Brazil
                [11 ]Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
                [12 ]Institut de Parasitologie et de Pathologie Tropicale, EA7292, University of Strasbourg, University of Strasbourg, Strasbourg, France
                Case Western Reserve University School of Medicine, UNITED STATES
                Author notes

                I have read the journal's policy and the authors of this manuscript have the following competing interests: LFP is a share holder of Kyolab laboratories. He also has a contact with Amazonia Fitomedicamentos to develop the ingenol derivatives. This does not alter the authors' adherence to Plos Pathogens policies on sharing data and materials.

                Conceived and designed the experiments: CVL OR. Performed the experiments: GD AK SB KF AAA ND AM CV BVD JSG TC. Analyzed the data: GD AK SB KF FC CR OR CVL CS AB MM BMP. Contributed reagents/materials/analysis tools: LFP LG JV. Wrote the paper: GD AK CVL. Performed patient selection: KK NC SDW.

                ‡ OR and CVL also contributed equally to this work.

                Article
                PPATHOGENS-D-15-00502
                10.1371/journal.ppat.1005063
                4520688
                26225566
                bdd9eb66-d04a-4946-8cc8-1648e711f085
                Copyright @ 2015

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited

                History
                : 27 February 2015
                : 2 July 2015
                Page count
                Figures: 10, Tables: 0, Pages: 36
                Funding
                We acknowledge grant support from the “Agence Nationale de Recherches sur le SIDA” (ANRS, France), the Belgian Fund for Scientific Research (FRS-FNRS, Belgium), the “Fondation Roi Baudouin”, the NEAT program, the Walloon Region (the Excellence Program “Cibles”) and the “Institut Universitaire de France (IUF)” (to OR). AK is a post-doctoral fellow of "Les Amis des Instituts Pasteur à Bruxelles, asbl". SB is a fellow of the Belgian « Fonds pour la Recherche dans l’Industrie et l’Agriculture (FRIA) ». BVD is an ANRS post-doctoral fellow. GD and CVL are Aspirant fellow and Research Director of the FRS-FNRS, respectively. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Custom metadata
                All relevant data are within the paper and its Supporting Information files.

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article