301
|
Vincendeau M, Göttesdorfer I, Schreml JMH, Wetie AGN, Mayer J, Greenwood AD, Helfer M, Kramer S, Seifarth W, Hadian K, Brack-Werner R, Leib-Mösch C. Modulation of human endogenous retrovirus (HERV) transcription during persistent and de novo HIV-1 infection. Retrovirology 2015; 12:27. [PMID: 25886562 PMCID: PMC4375885 DOI: 10.1186/s12977-015-0156-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 03/05/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The human genome contains multiple LTR elements including human endogenous retroviruses (HERVs) that together account for approximately 8-9% of the genomic DNA. At least 40 different HERV groups have been assigned to three major HERV classes on the basis of their homologies to exogenous retroviruses. Although most HERVs are silenced by a variety of genetic and epigenetic mechanisms, they may be reactivated by environmental stimuli such as exogenous viruses and thus may contribute to pathogenic conditions. The objective of this study was to perform an in-depth analysis of the influence of HIV-1 infection on HERV activity in different cell types. RESULTS A retrovirus-specific microarray that covers major HERV groups from all three classes was used to analyze HERV transcription patterns in three persistently HIV-1 infected cell lines of different cellular origins and in their uninfected counterparts. All three persistently infected cell lines showed increased transcription of multiple class I and II HERV groups. Up-regulated transcription of five HERV taxa (HERV-E, HERV-T, HERV-K (HML-10) and two ERV9 subgroups) was confirmed by quantitative reverse transcriptase PCR analysis and could be reversed by knock-down of HIV-1 expression with HIV-1-specific siRNAs. Cells infected de novo by HIV-1 showed stronger transcriptional up-regulation of the HERV-K (HML-2) group than persistently infected cells of the same origin. Analysis of transcripts from individual members of this group revealed up-regulation of predominantly two proviral loci (ERVK-7 and ERVK-15) on chromosomes 1q22 and 7q34 in persistently infected KE37.1 cells, as well as in de novo HIV-1 infected LC5 cells, while only one single HML-2 locus (ERV-K6) on chromosome 7p22.1 was activated in persistently infected LC5 cells. CONCLUSIONS Our results demonstrate that HIV-1 can alter HERV transcription patterns of infected cells and indicate a correlation between activation of HERV elements and the level of HIV-1 production. Moreover, our results suggest that the effects of HIV-1 on HERV activity may be far more extensive and complex than anticipated from initial studies with clinical material.
Collapse
Affiliation(s)
- Michelle Vincendeau
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany. .,Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Ingmar Göttesdorfer
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Julia M H Schreml
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Armand G Ngounou Wetie
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Jens Mayer
- Department of Human Genetics, Center of Human and Molecular Biology, Medical Faculty, University of Saarland, Homburg, Germany.
| | - Alex D Greenwood
- Department of Wildlife Diseases, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany.
| | - Markus Helfer
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Susanne Kramer
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Wolfgang Seifarth
- Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Kamyar Hadian
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany. .,Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Ruth Brack-Werner
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Christine Leib-Mösch
- Institute of Virology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany. .,Department of Hematology and Oncology, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
302
|
Norton TD, Miller EA, Bhardwaj N, Landau NR. Vpx-containing dendritic cell vaccine induces CTLs and reactivates latent HIV-1 in vitro. Gene Ther 2015; 22:227-36. [PMID: 25567537 PMCID: PMC4698816 DOI: 10.1038/gt.2014.117] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/25/2014] [Accepted: 11/17/2014] [Indexed: 12/20/2022]
Abstract
Eradication of human immunodeficiency virus-1 (HIV-1) from an infected individual requires a means of inducing production of virus from latently infected cells and stimulating an immune response against the infected cells. We report the development of lentiviral vectors that transduce dendritic cells (DCs) to both induce production of virus from latently infected cells and stimulate antigen-specific cytotoxic T lymphocytes (CTLs). The vectors package Vpx, a lentiviral accessory protein that counteracts the SAMHD1-mediated block to DC transduction, allowing for long-term expression of vector-encoded proteins. The vectors encode influenza or HIV-1-derived epitopes fused via a self-cleaving peptide to CD40L that releases the peptide into the endoplasmic reticulum for entry into the antigen presentation pathway. Expression of CD40L caused transduced DCs to mature and produce Th1-skewing cytokines. The DCs presented antigen to CD8 T cells, enhancing antigen-specific CTLs. Coculture of the transduced DCs with latently infected cells induced high-level virus production, an effect that was mediated by tumor necrosis factor alpha. The ability of a DC vaccine to reactivate latent HIV-1 and stimulate an adaptive immune response provide a means to reduce the size of the latent reservoir in patients. This strategy can also be applied to develop DC vaccines for other diseases.
Collapse
Affiliation(s)
- Thomas D. Norton
- Department of Medicine, NYU School of Medicine; New York, NY
- Department of Microbiology, NYU School of Medicine; New York, NY
| | - Elizabeth A. Miller
- Department of Medicine, Icahn School of Medicine at Mount Sinai; New York, NY
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | |
Collapse
|
303
|
Abstract
Despite effective treatment, HIV is not completely eliminated from the infected organism because of the existence of viral reservoirs. A major reservoir consists of infected resting CD4+ T cells, mostly of memory type, that persist over time due to the stable proviral insertion and a long cellular lifespan. Resting cells do not produce viral particles and are protected from viral-induced cytotoxicity or immune killing. However, these latently infected cells can be reactivated by stochastic events or by external stimuli. The present review focuses on novel genome-wide technologies applied to the study of integration, transcriptome, and proteome characteristics and their recent contribution to the understanding of HIV latency.
Collapse
Affiliation(s)
- Angela Ciuffi
- Institute of Microbiology, University Hospital of Lausanne (CHUV), University of Lausanne, Bugnon 48, 1011, Lausanne, Switzerland,
| | | | | | | | | |
Collapse
|
304
|
Zhu W, Lei R, Le Duff Y, Li J, Guo F, Wainberg MA, Liang C. The CRISPR/Cas9 system inactivates latent HIV-1 proviral DNA. Retrovirology 2015; 12:22. [PMID: 25808449 PMCID: PMC4359768 DOI: 10.1186/s12977-015-0150-z] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/09/2015] [Indexed: 12/18/2022] Open
Abstract
Background Highly active antiretroviral therapy (HAART) has transformed HIV-1 infection from a deadly disease to a manageable chronic illness, albeit does not provide a cure. The recently developed genome editing system called CRISPR/Cas9 offers a new tool to inactivate the integrated latent HIV-1 DNA and may serve as a new avenue toward cure. Findings We tested 10 sites in HIV-1 DNA that can be targeted by CRISPR/Cas9. The engineered CRISPR/Cas9 system was introduced into the JLat10.6 cells that are latently infected by HIV-1. The sequencing results showed that each target site in HIV-1 DNA was efficiently mutated by CRISPR/Cas9 with the target site in the second exon of Rev (called T10) exhibiting the highest degree of mutation. As a result, HIV-1 gene expression and virus production were significantly diminished with T10 causing a 20-fold reduction. Conclusions The CRISPR/Cas9 complex efficiently mutates and deactivates HIV-1 proviral DNA in latently infected Jurkat cells. Our results also revealed a highly efficient Cas9 target site within the second exon of Rev that represents a promising target to be further explored in the CRISPR/Cas9-based cure strategy.
Collapse
|
305
|
Imami N, Herasimtschuk AA. Multifarious immunotherapeutic approaches to cure HIV-1 infection. Hum Vaccin Immunother 2015; 11:2287-93. [PMID: 26048144 PMCID: PMC4635699 DOI: 10.1080/21645515.2015.1021523] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/15/2015] [Indexed: 01/19/2023] Open
Abstract
Immunotherapy in the context of treated HIV-1 infection aims to improve immune responses to achieve better control of the virus. To date, multifaceted immunotherapeutic approaches have been shown to reduce immune activation and increase CD4 T-lymphocyte counts, further to the effects of antiretroviral therapy alone, in addition to improving HIV-1-specific T-cell responses. While sterilizing cure of HIV-1 would involve elimination of all replication-competent virus, a functional cure in which the host has long-lasting control of viral replication may be more feasible. In this commentary, we discuss novel strategies aimed at targeting the latent viral reservoir with cure of HIV-1 infection being the ultimate goal, an achievement that would have considerable impact on worldwide HIV-1 infection.
Collapse
Affiliation(s)
- Nesrina Imami
- Department of Medicine; Imperial College London; London, UK
| | | |
Collapse
|
306
|
Anderson JL, Fromentin R, Corbelli GM, Østergaard L, Ross AL. Progress towards an HIV cure: update from the 2014 International AIDS Society Symposium. AIDS Res Hum Retroviruses 2015; 31:36-44. [PMID: 25257573 DOI: 10.1089/aid.2014.0236] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Biomedical research has led to profound advances in the treatment of HIV infection. Combination antiretroviral therapy (ART) now provides the means to readily control viral infection, and people living with HIV who receive timely and effective ART can expect to benefit from a life expectancy comparable to uninfected individuals. Nevertheless, despite effective treatment, ART does not fully restore the immune system and importantly HIV persists indefinitely in latent reservoirs, resulting in the need for life-long treatment. The challenges and limits of life-long treatment have spurred significant scientific interest and global investment into research towards an HIV cure. The International AIDS Society (IAS) 2014 Towards an HIV cure symposium brought together researchers and community to discuss the most recent advances in our understanding of latency and HIV reservoirs, and the clinical approaches towards an HIV cure under current investigation. This report summarizes and reviews some of the major findings discussed during the symposium.
Collapse
Affiliation(s)
| | - Rémi Fromentin
- Vaccine and Gene Therapy Institute Florida, Port St. Lucie, Florida
| | | | | | - Anna Laura Ross
- ANRS, Paris, France
- International AIDS Society, Geneva, Switzerland
| |
Collapse
|
307
|
Miller EA, Spadaccia MR, Norton T, Demmler M, Gopal R, O'Brien M, Landau N, Dubensky TW, Lauer P, Brockstedt DG, Bhardwaj N. Attenuated Listeria monocytogenes vectors overcome suppressive plasma factors during HIV infection to stimulate myeloid dendritic cells to promote adaptive immunity and reactivation of latent virus. AIDS Res Hum Retroviruses 2015; 31:127-36. [PMID: 25376024 PMCID: PMC4287309 DOI: 10.1089/aid.2014.0138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
HIV-1 infection is characterized by myeloid dendritic cell (DC) dysfunction, which blunts the responsiveness to vaccine adjuvants. We previously showed that nonviral factors in HIV-seropositive plasma are partially responsible for mediating this immune suppression. In this study we investigated recombinant Listeria monocytogenes (Lm) vectors, which naturally infect and potently activate DCs from seronegative donors, as a means to overcome DC dysfunction associated with HIV infection. Monocyte-derived DCs were cocultured with plasma from HIV-infected donors (HIV-moDCs) to induce a dysregulated state and infected with an attenuated, nonreplicative vaccine strain of Lm expressing full length clade B consensus gag (KBMA Lm-gag). Lm infection stimulated cytokine secretion [interleukin (IL)-12p70, tumor necrosis factor (TNF)-α, and IL-6] and Th-1 skewing of allogeneic naive CD4 T cells by HIV-moDCs, in contrast to the suppressive effects observed by HIV plasma on moDCs on toll-like receptor ligand stimulation. Upon coculture of "killed" but metabolically active (KBMA) Lm-gag-infected moDCs from HIV-infected donors with autologous cells, expansion of polyfunctional, gag-specific CD8(+) T cells was observed. Reactivation of latent proviruses by moDCs following Lm infection was also observed in models of HIV latency in a TNF-α-dependent manner. These findings reveal the unique ability of Lm vectors to contend with dysregulation of HIV-moDCs, while simultaneously possessing the capacity to activate latent virus. Concurrent stimulation of innate and adaptive immunity and disruption of latency may be an approach to reduce the pool of latently infected cells during HIV infection. Further study of Lm vectors as part of therapeutic vaccination and eradication strategies may advance this evolving field.
Collapse
Affiliation(s)
- Elizabeth A. Miller
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Thomas Norton
- Division of Infectious Diseases, New York University School of Medicine, New York, New York
| | - Morgan Demmler
- Cancer Institute, New York University School of Medicine, New York, New York
| | - Ramya Gopal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Meagan O'Brien
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nathaniel Landau
- Department of Microbiology, New York University School of Medicine, New York, New York
| | | | | | | | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
308
|
Jiang G, Dandekar S. Targeting NF-κB signaling with protein kinase C agonists as an emerging strategy for combating HIV latency. AIDS Res Hum Retroviruses 2015; 31:4-12. [PMID: 25287643 DOI: 10.1089/aid.2014.0199] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Highly active antiretroviral therapy (HAART) is very effective in suppressing HIV-1 replication and restoring immune functions in HIV-infected individuals. However, it fails to eradicate the latent viral reservoirs and fully resolve chronic inflammation in HIV infection. The "shock-and-kill" strategy was recently proposed to induce latent HIV expression in the presence of HAART. Recent studies have shown that the protein kinase C (PKC) agonists are highly potent in inducing latent HIV expression from the viral reservoirs in vitro and ex vivo and in protecting primary CD4(+) T cells from HIV infection through down-modulation of their HIV coreceptor expression. The PKC agonists are excellent candidates for advancing to clinical HIV eradication strategies. This article will present a critical review of the structure and function of known PKC agonists, their mechanisms for the reactivation of latent HIV expression, and the potential of these compounds for advancing clinical HIV eradication strategies.
Collapse
Affiliation(s)
- Guochun Jiang
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - Satya Dandekar
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| |
Collapse
|
309
|
Craigie R, Bushman FD. Host Factors in Retroviral Integration and the Selection of Integration Target Sites. Microbiol Spectr 2014; 2:10.1128/microbiolspec.MDNA3-0026-2014. [PMID: 26104434 PMCID: PMC4525071 DOI: 10.1128/microbiolspec.mdna3-0026-2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Indexed: 02/07/2023] Open
Abstract
In order to replicate, a retrovirus must integrate a DNA copy of the viral RNA genome into a chromosome of the host cell. The study of retroviral integration has advanced considerably in the past few years. Here we focus on host factor interactions and the linked area of integration targeting. Genome-wide screens for cellular factors affecting HIV replication have identified a series of host cell proteins that may mediate subcellular trafficking for preintegration complexes, nuclear import, and integration target site selection. The cell transcriptional co-activator protein LEDGF/p75 has been identified as a tethering factor important for HIV integration, and recently, BET proteins (Brd2, 4, and 4) have been identified as tethering factors for the gammaretroviruses. A new class of HIV inhibitors has been developed targeting the HIV-1 IN-LEDGF binding site, though surprisingly these inhibitors appear to block assembly late during replication and do not act at the integration step. Going forward, genome-wide studies of HIV-host interactions offer many new starting points to investigate HIV replication and identify potential new inhibitor targets.
Collapse
Affiliation(s)
- Robert Craigie
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0560
| | - Frederic D. Bushman
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
310
|
Blankson JN, Siliciano JD, Siliciano RF. Finding a cure for human immunodeficiency virus-1 infection. Infect Dis Clin North Am 2014; 28:633-50. [PMID: 25277513 PMCID: PMC4253590 DOI: 10.1016/j.idc.2014.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Remarkable advances have been made in the treatment of human immunodeficiency virus (HIV)-1 infection, but in the entire history of the epidemic, only 1 patient has been cured. Herein we review the fundamental mechanisms that render HIV-1 infection difficult to cure and then discuss recent clinical and experimental situations in which some form of cure has been achieved. Finally, we consider approaches that are currently being taken to develop a general cure for HIV-1 infection.
Collapse
Affiliation(s)
- Joel N Blankson
- Department of Medicine, Johns Hopkins University School of Medicine, 733, North Broadway, Baltimore, MD 21205, USA
| | - Janet D Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, 733, North Broadway, Baltimore, MD 21205, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, 733, North Broadway, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, 733, North Broadway, Baltimore, MD 21205, USA.
| |
Collapse
|
311
|
De Crignis E, Mahmoudi T. HIV eradication: combinatorial approaches to activate latent viruses. Viruses 2014; 6:4581-608. [PMID: 25421889 PMCID: PMC4246239 DOI: 10.3390/v6114581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/01/2014] [Accepted: 11/13/2014] [Indexed: 12/11/2022] Open
Abstract
The concept of eradication of the Human Immune Deficiency Virus (HIV) from infected patients has gained much attention in the last few years. While combination Anti-Retroviral Therapy (c-ART) has been extremely effective in suppressing viral replication, it is not curative. This is due to the presence of a reservoir of latent HIV infected cells, which persist in the presence of c-ART. Recently, pharmaceutical approaches have focused on the development of molecules able to induce HIV-1 replication from latently infected cells in order to render them susceptible to viral cytopathic effects and host immune responses. Alternative pathways and transcription complexes function to regulate the activity of the HIV promoter and might serve as molecular targets for compounds to activate latent HIV. A combined therapy coupling various depressors and activators will likely be the most effective in promoting HIV replication while avoiding pleiotropic effects at the cellular level. Moreover, in light of differences among HIV subtypes and variability in integration sites, the combination of multiple agents targeting multiple pathways will increase likelihood of therapeutic effectiveness and prevent mutational escape. This review provides an overview of the mechanisms that can be targeted to induce HIV activation focusing on potential combinatorial approaches.
Collapse
Affiliation(s)
- Elisa De Crignis
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam 3015 CN, The Netherlands.
| |
Collapse
|
312
|
Lu HK, Gray LR, Wightman F, Ellenberg P, Khoury G, Cheng WJ, Mota TM, Wesselingh S, Gorry PR, Cameron PU, Churchill MJ, Lewin SR. Ex vivo response to histone deacetylase (HDAC) inhibitors of the HIV long terminal repeat (LTR) derived from HIV-infected patients on antiretroviral therapy. PLoS One 2014; 9:e113341. [PMID: 25409334 PMCID: PMC4237424 DOI: 10.1371/journal.pone.0113341] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/22/2014] [Indexed: 01/31/2023] Open
Abstract
Histone deacetylase inhibitors (HDACi) can induce human immunodeficiency virus (HIV) transcription from the HIV long terminal repeat (LTR). However, ex vivo and in vivo responses to HDACi are variable and the activity of HDACi in cells other than T-cells have not been well characterised. Here, we developed a novel assay to determine the activity of HDACi on patient-derived HIV LTRs in different cell types. HIV LTRs from integrated virus were amplified using triple-nested Alu-PCR from total memory CD4+ T-cells (CD45RO+) isolated from HIV-infected patients prior to and following suppressive antiretroviral therapy. NL4-3 or patient-derived HIV LTRs were cloned into the chromatin forming episomal vector pCEP4, and the effect of HDACi investigated in the astrocyte and epithelial cell lines SVG and HeLa, respectively. There were no significant differences in the sequence of the HIV LTRs isolated from CD4+ T-cells prior to and after 18 months of combination antiretroviral therapy (cART). We found that in both cell lines, the HDACi panobinostat, trichostatin A, vorinostat and entinostat activated patient-derived HIV LTRs to similar levels seen with NL4-3 and all patient derived isolates had similar sensitivity to maximum HDACi stimulation. We observed a marked difference in the maximum fold induction of luciferase by HDACi in HeLa and SVG, suggesting that the effect of HDACi may be influenced by the cellular environment. Finally, we observed significant synergy in activation of the LTR with vorinostat and the viral protein Tat. Together, our results suggest that the LTR sequence of integrated virus is not a major determinant of a functional response to an HDACi.
Collapse
Affiliation(s)
- Hao K. Lu
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - Lachlan R. Gray
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - Fiona Wightman
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - Paula Ellenberg
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - Gabriela Khoury
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - Wan-Jung Cheng
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - Talia M. Mota
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Victoria, Australia
| | - Steve Wesselingh
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Paul R. Gorry
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Victoria, Australia
| | - Paul U. Cameron
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
- Infectious Disease Unit, Alfred Hospital, Melbourne, Victoria, Australia
| | - Melissa J. Churchill
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
- Department of Medicine, Monash University, Clayton, Victoria, Australia
| | - Sharon R. Lewin
- Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
- Infectious Disease Unit, Alfred Hospital, Melbourne, Victoria, Australia
- Peter Doherty Institute, Melbourne University, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
313
|
Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS Pathog 2014; 10:e1004473. [PMID: 25393648 PMCID: PMC4231123 DOI: 10.1371/journal.ppat.1004473] [Citation(s) in RCA: 416] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/15/2014] [Indexed: 02/06/2023] Open
Abstract
Human immunodeficiency virus (HIV) persistence in latently infected resting memory CD4+ T-cells is the major barrier to HIV cure. Cellular histone deacetylases (HDACs) are important in maintaining HIV latency and histone deacetylase inhibitors (HDACi) may reverse latency by activating HIV transcription from latently infected CD4+ T-cells. We performed a single arm, open label, proof-of-concept study in which vorinostat, a pan-HDACi, was administered 400 mg orally once daily for 14 days to 20 HIV-infected individuals on suppressive antiretroviral therapy (ART). The primary endpoint was change in cell associated unspliced (CA-US) HIV RNA in total CD4+ T-cells from blood at day 14. The study is registered at ClinicalTrials.gov (NCT01365065). Vorinostat was safe and well tolerated and there were no dose modifications or study drug discontinuations. CA-US HIV RNA in blood increased significantly in 18/20 patients (90%) with a median fold change from baseline to peak value of 7.4 (IQR 3.4, 9.1). CA-US RNA was significantly elevated 8 hours post drug and remained elevated 70 days after last dose. Significant early changes in expression of genes associated with chromatin remodeling and activation of HIV transcription correlated with the magnitude of increased CA-US HIV RNA. There were no statistically significant changes in plasma HIV RNA, concentration of HIV DNA, integrated DNA, inducible virus in CD4+ T-cells or markers of T-cell activation. Vorinostat induced a significant and sustained increase in HIV transcription from latency in the majority of HIV-infected patients. However, additional interventions will be needed to efficiently induce virus production and ultimately eliminate latently infected cells. Trial Registration ClinicalTrials.gov NCT01365065 The major barrier to curing HIV is the long term persistence of latently infected resting memory T-cells in HIV-infected patients on antiretroviral therapy (ART). One strategy being pursued to eliminate latently infected cells is to activate HIV production from latently infected cells with the aim of killing latently infected cells via virus induced cell death or stimulation of an HIV-specific immune response. Histone deacetylases (HDACs) are important in maintaining HIV latency. Vorinostat, an inhibitor of HDACs (HDACi) licensed for the treatment of some malignancies, has been shown in laboratory studies and a clinical study of selected individuals to disrupt HIV latency. We examined the ability of standard dose vorinostat given daily for 14 days to activate latent HIV infection in unselected HIV-infected individuals on ART. The study showed evidence of activation of latent HIV infection in 18/20 (90%) of individuals and was safe and generally well tolerated. There were significant early changes in host gene expression, which persisted during and after the period of vorinostat. No changes were seen in immune activation or number of latently infected cells. Vorinostat was able to activate latent HIV infection in most individuals. Additional interventions will be needed to eliminate latent HIV infection.
Collapse
|
314
|
Kim M, Hosmane NN, Bullen CK, Capoferri A, Yang HC, Siliciano JD, Siliciano RF. A primary CD4(+) T cell model of HIV-1 latency established after activation through the T cell receptor and subsequent return to quiescence. Nat Protoc 2014; 9:2755-70. [PMID: 25375990 DOI: 10.1038/nprot.2014.188] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A mechanistic understanding of HIV-1 latency depends on a model system that recapitulates the in vivo condition of latently infected, resting CD4(+) T lymphocytes. Latency seems to be established after activated CD4(+) T cells, the principal targets of HIV-1 infection, become productively infected and survive long enough to return to a resting memory state in which viral expression is inhibited by changes in the cellular environment. This protocol describes an ex vivo primary cell system that is generated under conditions that reflect the in vivo establishment of latency. Creation of these latency model cells takes 12 weeks and, once established, the cells can be maintained and used for several months. The resulting cell population contains both uninfected and latently infected cells. This primary cell model can be used to perform drug screens, to study cytolytic T lymphocyte (CTL) responses to HIV-1, to compare viral alleles or to expand the ex vivo life span of cells from HIV-1-infected individuals for extended study.
Collapse
Affiliation(s)
- Michelle Kim
- 1] Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. [2] Howard Hughes Medical Institute, Baltimore, Maryland, USA
| | - Nina N Hosmane
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C Korin Bullen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adam Capoferri
- 1] Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. [2] Howard Hughes Medical Institute, Baltimore, Maryland, USA
| | - Hung-Chih Yang
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Janet D Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert F Siliciano
- 1] Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. [2] Howard Hughes Medical Institute, Baltimore, Maryland, USA
| |
Collapse
|
315
|
Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol 2014; 12:750-64. [PMID: 25402363 PMCID: PMC4383747 DOI: 10.1038/nrmicro3352] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Effective antiretroviral therapy (ART) blunts viraemia, which enables HIV-1-infected individuals to control infection and live long, productive lives. However, HIV-1 infection remains incurable owing to the persistence of a viral reservoir that harbours integrated provirus within host cellular DNA. This latent infection is unaffected by ART and hidden from the immune system. Recent studies have focused on the development of therapies to disrupt latency. These efforts unmasked residual viral genomes and highlighted the need to enable the clearance of latently infected cells, perhaps via old and new strategies that improve the HIV-1-specific immune response. In this Review, we explore new approaches to eradicate established HIV-1 infection and avoid the burden of lifelong ART.
Collapse
Affiliation(s)
- Nancie M Archin
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Julia Marsh Sung
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Carolina Garrido
- Department of Medicine, University of North Carolina at Chapel Hill
| | | | - David M Margolis
- 1] Department of Medicine, University of North Carolina at Chapel Hill. [2] Department of Microbiology and Immunology, University of North Carolina at Chapel Hill. [3] Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
316
|
Murry JP, Godoy J, Mukim A, Swann J, Bruce JW, Ahlquist P, Bosque A, Planelles V, Spina CA, Young JAT. Sulfonation pathway inhibitors block reactivation of latent HIV-1. Virology 2014; 471-473:1-12. [PMID: 25310595 DOI: 10.1016/j.virol.2014.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 06/16/2014] [Accepted: 08/18/2014] [Indexed: 12/12/2022]
Abstract
Long-lived pools of latently infected cells are a significant barrier to the development of a cure for HIV-1 infection. A better understanding of the mechanisms of reactivation from latency is needed to facilitate the development of novel therapies that address this problem. Here we show that chemical inhibitors of the sulfonation pathway prevent virus reactivation, both in latently infected J-Lat and U1 cell lines and in a primary human CD4+ T cell model of latency. In each of these models, sulfonation inhibitors decreased transcription initiation from the HIV-1 promoter. These inhibitors block transcription initiation at a step that lies downstream of nucleosome remodeling and affects RNA polymerase II recruitment to the viral promoter. These results suggest that the sulfonation pathway acts by a novel mechanism to regulate efficient virus transcription initiation during reactivation from latency, and further that augmentation of this pathway could be therapeutically useful.
Collapse
Affiliation(s)
- Jeffrey P Murry
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joseph Godoy
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Amey Mukim
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Justine Swann
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - James W Bruce
- Morgridge Institute for Research, Madison, WI, USA; Institute for Molecular Virology, University of Wisconsin, Madison, WI, USA; McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Paul Ahlquist
- Morgridge Institute for Research, Madison, WI, USA; Institute for Molecular Virology, University of Wisconsin, Madison, WI, USA; McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Alberto Bosque
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Vicente Planelles
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Celsa A Spina
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - John A T Young
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA; Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
317
|
Andrews IP, Ketcham JM, Blumberg PM, Kedei N, Lewin N, Peach ML, Krische MJ. Synthesis of seco-B-ring bryostatin analogue WN-1 via C-C bond-forming hydrogenation: critical contribution of the B-ring in determining bryostatin-like and phorbol 12-myristate 13-acetate-like properties. J Am Chem Soc 2014; 136:13209-16. [PMID: 25207655 PMCID: PMC4183601 DOI: 10.1021/ja507825s] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Indexed: 01/31/2023]
Abstract
The seco-B-ring bryostatin analogue, macrodiolide WN-1, was prepared in 17 steps (longest linear sequence) and 30 total steps with three bonds formed via hydrogen-mediated C-C coupling. This synthetic route features a palladium-catalyzed alkoxycarbonylation of a C2-symmetric diol to form the C9-deoxygenated bryostatin A-ring. WN-1 binds to PKCα (Ki = 16.1 nM) and inhibits the growth of multiple leukemia cell lines. Although structural features of the WN-1 A-ring and C-ring are shared by analogues that display bryostatin-like behavior, WN-1 displays PMA-like behavior in U937 cell attachment and proliferation assays, as well as in K562 and MV-4-11 proliferation assays. Molecular modeling studies suggest the pattern of internal hydrogen bonds evident in bryostatin 1 is preserved in WN-1, and that upon docking WN-1 into the crystal structure of the C1b domain of PKCδ, the binding mode of bryostatin 1 is reproduced. The collective data emphasize the critical contribution of the B-ring to the function of the upper portion of the molecule in conferring a bryostatin-like pattern of biological activity.
Collapse
Affiliation(s)
- Ian P. Andrews
- Department
of Chemistry and Biochemistry, University
of Texas at Austin, Austin, Texas 78712, United States
| | - John M. Ketcham
- Department
of Chemistry and Biochemistry, University
of Texas at Austin, Austin, Texas 78712, United States
| | - Peter M. Blumberg
- Laboratory
of Cancer Biology and Genetics, National
Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, United States
| | - Noemi Kedei
- Laboratory
of Cancer Biology and Genetics, National
Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, United States
| | - Nancy
E. Lewin
- Laboratory
of Cancer Biology and Genetics, National
Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, United States
| | - Megan L. Peach
- Basic Science Program,
Leidos Biomedical Research, Inc., Chemical Biology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Michael J. Krische
- Department
of Chemistry and Biochemistry, University
of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
318
|
Stevenson M. Role of myeloid cells in HIV-1-host interplay. J Neurovirol 2014; 21:242-8. [PMID: 25236811 DOI: 10.1007/s13365-014-0281-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/25/2014] [Accepted: 08/14/2014] [Indexed: 12/11/2022]
Abstract
The AIDS research field has embarked on a bold mission to cure HIV-1-infected individuals of the virus. To do so, scientists are attempting to identify the reservoirs that support viral persistence in patients on therapy, to understand how viral persistence is regulated and to come up with strategies that interrupt viral persistence and that eliminate the viral reservoirs. Most of the attention regarding the cure of HIV-1 infection has focused on the CD4+ T cell reservoir. Investigators are developing tools to probe the CD4+ T cell reservoirs as well as in vitro systems that provide clues on how to perturb them. By comparison, the myeloid cell, and in particular, the macrophage has received far less attention. As a consequence, there is very little understanding as to the role played by myeloid cells in viral persistence in HIV-1-infected individuals on suppressive therapy. As such, should myeloid cells constitute a viral reservoir, unique strategies may be required for their elimination. This article will overview research that is examining the role of macrophage in virus-host interplay and will discuss features of this interplay that could impact efforts to eliminate myeloid cell reservoirs.
Collapse
Affiliation(s)
- Mario Stevenson
- Department of Medicine, University of Miami Medical School, Miami, FL, USA,
| |
Collapse
|
319
|
Cornet S, Nicot A, Rivero A, Gandon S. Evolution of Plastic Transmission Strategies in Avian Malaria. PLoS Pathog 2014; 10:e1004308. [PMID: 25210974 PMCID: PMC4161439 DOI: 10.1371/journal.ppat.1004308] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 07/02/2014] [Indexed: 01/26/2023] Open
Abstract
Malaria parasites have been shown to adjust their life history traits to changing environmental conditions. Parasite relapses and recrudescences—marked increases in blood parasite numbers following a period when the parasite was either absent or present at very low levels in the blood, respectively—are expected to be part of such adaptive plastic strategies. Here, we first present a theoretical model that analyses the evolution of transmission strategies in fluctuating seasonal environments and we show that relapses may be adaptive if they are concomitant with the presence of mosquitoes in the vicinity of the host. We then experimentally test the hypothesis that Plasmodium parasites can respond to the presence of vectors. For this purpose, we repeatedly exposed birds infected by the avian malaria parasite Plasmodium relictum to the bites of uninfected females of its natural vector, the mosquito Culex pipiens, at three different stages of the infection: acute (∼34 days post infection), early chronic (∼122 dpi) and late chronic (∼291 dpi). We show that: (i) mosquito-exposed birds have significantly higher blood parasitaemia than control unexposed birds during the chronic stages of the infection and that (ii) this translates into significantly higher infection prevalence in the mosquito. Our results demonstrate the ability of Plasmodium relictum to maximize their transmission by adopting plastic life history strategies in response to the availability of insect vectors. Seasonal fluctuations in the environment affect dramatically the abundance of insect species. These fluctuations have important consequences for the transmission of vector-borne diseases. Here we contend that malaria parasites may have evolved plastic transmission strategies as an adaptation to the fluctuations in mosquito densities. First, our theoretical analysis identifies the conditions for the evolution of such plastic transmission strategies. Second, we show that in avian malaria Plasmodium parasites have the ability to increase transmission after being bitten by uninfected Culex mosquitoes. This demonstrates the ability of Plasmodium parasites to adopt plastic transmission strategies and challenges our understanding of malaria epidemiology.
Collapse
Affiliation(s)
- Stéphane Cornet
- Centre d'Ecologie Fonctionnelle et Evolutive (CEFE), UMR CNRS 5175 - Université de Montpellier - Université Paul-Valéry Montpellier - EPHE, Montpellier, France
- Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UMR CNRS 5290-IRD 224-UM1-UM2, Montpellier, France
| | - Antoine Nicot
- Centre d'Ecologie Fonctionnelle et Evolutive (CEFE), UMR CNRS 5175 - Université de Montpellier - Université Paul-Valéry Montpellier - EPHE, Montpellier, France
- Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UMR CNRS 5290-IRD 224-UM1-UM2, Montpellier, France
| | - Ana Rivero
- Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UMR CNRS 5290-IRD 224-UM1-UM2, Montpellier, France
| | - Sylvain Gandon
- Centre d'Ecologie Fonctionnelle et Evolutive (CEFE), UMR CNRS 5175 - Université de Montpellier - Université Paul-Valéry Montpellier - EPHE, Montpellier, France
- * E-mail:
| |
Collapse
|
320
|
Sebastian NT, Collins KL. Targeting HIV latency: resting memory T cells, hematopoietic progenitor cells and future directions. Expert Rev Anti Infect Ther 2014; 12:1187-201. [PMID: 25189526 DOI: 10.1586/14787210.2014.956094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Current therapy for HIV effectively suppresses viral replication and prolongs life, but the infection persists due, at least in part, to latent infection of long-lived cells. One favored strategy toward a cure targets latent virus in resting memory CD4(+) T cells by stimulating viral production. However, the existence of an additional reservoir in bone marrow hematopoietic progenitor cells has been detected in some treated HIV-infected people. This review describes approaches investigators have used to reactivate latent proviral genomes in resting CD4(+) T cells and hematopoietic progenitor cells. In addition, the authors review approaches for clearance of these reservoirs along with other important topics related to HIV eradication.
Collapse
Affiliation(s)
- Nadia T Sebastian
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
321
|
Selective HDAC inhibition for the disruption of latent HIV-1 infection. PLoS One 2014; 9:e102684. [PMID: 25136952 PMCID: PMC4138023 DOI: 10.1371/journal.pone.0102684] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 06/23/2014] [Indexed: 11/30/2022] Open
Abstract
Selective histone deacetylase (HDAC) inhibitors have emerged as a potential anti-latency therapy for persistent human immunodeficiency virus type 1 (HIV-1) infection. We utilized a combination of small molecule inhibitors and short hairpin (sh)RNA-mediated gene knockdown strategies to delineate the key HDAC(s) to be targeted for selective induction of latent HIV-1 expression. Individual depletion of HDAC3 significantly induced expression from the HIV-1 promoter in the 2D10 latency cell line model. However, depletion of HDAC1 or −2 alone or in combination did not significantly induce HIV-1 expression. Co-depletion of HDAC2 and −3 resulted in a significant increase in expression from the HIV-1 promoter. Furthermore, concurrent knockdown of HDAC1, −2, and −3 resulted in a significant increase in expression from the HIV-1 promoter. Using small molecule HDAC inhibitors of differing selectivity to ablate the residual HDAC activity that remained after (sh)RNA depletion, the effect of depletion of HDAC3 was further enhanced. Enzymatic inhibition of HDAC3 with the selective small-molecule inhibitor BRD3308 activated HIV-1 transcription in the 2D10 cell line. Furthermore, ex vivo exposure to BRD3308 induced outgrowth of HIV-1 from resting CD4+ T cells isolated from antiretroviral-treated, aviremic HIV+ patients. Taken together these findings suggest that HDAC3 is an essential target to disrupt HIV-1 latency, and inhibition of HDAC2 may also contribute to the effort to purge and eradicate latent HIV-1 infection.
Collapse
|
322
|
Garrido C, Margolis DM. Translational challenges in targeting latent HIV infection and the CNS reservoir problem. J Neurovirol 2014; 21:222-6. [PMID: 25060298 DOI: 10.1007/s13365-014-0269-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/09/2014] [Accepted: 06/18/2014] [Indexed: 01/19/2023]
Abstract
Too controversial to discuss only a short time ago, achieving a cure for HIV infection has become a priority in HIV research. However, substantial challenges must be overcome. Among key hurdles to be surmounted is the definition of a reliable, validated model in which to test latency reversal agents (LRAs), as current primary cell models differ in their response to such agents. Animal models such as the HIV-infected humanized BLT mouse and SIV-infected macaque will be essential to study LRAs and to quantify their effects in anatomic reservoirs. Of several potential anatomic reservoirs, the central nervous system presents a significant obstacle, as it is known to harbor persistent HIV infection and is difficult to access for study and therapeutic intervention.
Collapse
Affiliation(s)
- Carolina Garrido
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | |
Collapse
|
323
|
Affiliation(s)
- Sharon R Lewin
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, VIC 3004, Australia; Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.
| | - Steven G Deeks
- Department of Medicine, San Francisco General Hospital, University California, San Francisco, San Francisco, CA, USA
| | - Françoise Barré-Sinoussi
- Inserm and Unit of Regulation of Retroviral Infections, Department of Virology, Institut Pasteur, Paris, France
| |
Collapse
|
324
|
Reactivation of HIV latency by a newly modified Ingenol derivative via protein kinase Cδ-NF-κB signaling. AIDS 2014; 28:1555-66. [PMID: 24804860 DOI: 10.1097/qad.0000000000000289] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Although HAART effectively suppresses viral replication, it fails to eradicate latent viral reservoirs. The 'shock and kill' strategy involves the activation of HIV from latent reservoirs and targeting them for eradication. Our goal was to develop new approaches for activating HIV from latent reservoirs. DESIGN We investigated capacity of Ingenol B (IngB), a newly modified derivative of Ingenol ester that was originally isolated from a Brazilian plant in Amazon, for its capacity and mechanisms of HIV reactivation. METHODS Reactivation of HIV-1 by IngB was evaluated in J-Lat A1 cell culture model of HIV latency as well as in purified primary CD4 T cells from long-term HAART-treated virologically-suppressed HIV-infected individuals. The underlining molecular mechanisms of viral reactivation were investigated using flow cytometry, RT-qPCR and chromatin immunoprecipitation. RESULTS IngB is highly effective in reactivating HIV in J-Lat A1 cells with relatively low cellular toxicity. It is also able to reactivate latent HIV in purified CD4 T cells from HAART-treated HIV-positive individuals ex vivo. Our data show that IngB may reactivate HIV expression by both activating protein kinase C (PKC)δ-nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway and directly inducing NF-κB protein expression. Importantly, IngB has a synergistic effect with JQ1, a BET bromodomain inhibitor, in latent HIV reactivation. CONCLUSIONS IngB is a new promising compound to activate latent HIV reservoirs. Our data suggest that formulating novel derivatives from Ingenol esters may be an innovative approach to develop new lead compounds to reactivate latent HIV.
Collapse
|
325
|
Siliciano JD, Siliciano RF. Recent developments in the search for a cure for HIV-1 infection: targeting the latent reservoir for HIV-1. J Allergy Clin Immunol 2014; 134:12-9. [PMID: 25117799 DOI: 10.1016/j.jaci.2014.05.026] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 02/07/2023]
Abstract
HIV-1 infection can now be readily controlled with combination antiretroviral therapy. However, the virus persists indefinitely in a stable latent reservoir in resting CD4(+) T cells. This reservoir generally prevents cure of the infection with combination antiretroviral therapy alone. However, several recent cases of potential HIV-1 cure have generated renewed optimism. Here we review these cases and consider new developments in our understanding of the latent reservoir. In addition, we consider clinical aspects of curative strategies to provide a more realistic picture of what a generally applicable cure for HIV-1 infection is likely to entail.
Collapse
Affiliation(s)
- Janet D Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md; Howard Hughes Medical Institute, Baltimore, Md.
| |
Collapse
|
326
|
Autran B, Hamimi C, Katlama C. One Step Closer to HIV Eradication? CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2014. [DOI: 10.1007/s40506-014-0017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
327
|
Mohammadi P, di Iulio J, Muñoz M, Martinez R, Bartha I, Cavassini M, Thorball C, Fellay J, Beerenwinkel N, Ciuffi A, Telenti A. Dynamics of HIV latency and reactivation in a primary CD4+ T cell model. PLoS Pathog 2014; 10:e1004156. [PMID: 24875931 PMCID: PMC4038609 DOI: 10.1371/journal.ppat.1004156] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/18/2014] [Indexed: 12/11/2022] Open
Abstract
HIV latency is a major obstacle to curing infection. Current strategies to eradicate HIV aim at increasing transcription of the latent provirus. In the present study we observed that latently infected CD4+ T cells from HIV-infected individuals failed to produce viral particles upon ex vivo exposure to SAHA (vorinostat), despite effective inhibition of histone deacetylases. To identify steps that were not susceptible to the action of SAHA or other latency reverting agents, we used a primary CD4+ T cell model, joint host and viral RNA sequencing, and a viral-encoded reporter. This model served to investigate the characteristics of latently infected cells, the dynamics of HIV latency, and the process of reactivation induced by various stimuli. During latency, we observed persistence of viral transcripts but only limited viral translation. Similarly, the reactivating agents SAHA and disulfiram successfully increased viral transcription, but failed to effectively enhance viral translation, mirroring the ex vivo data. This study highlights the importance of post-transcriptional blocks as one mechanism leading to HIV latency that needs to be relieved in order to purge the viral reservoir. HIV-infected individuals must receive lifelong antiviral therapy because treatment discontinuation generally results in rapid viral rebound. The field has identified a state of latency at the level of transcription of the integrated provirus as the major mechanism of persistence. A number of drugs are now tested that aim at inducing viral transcription as a step to purge the reservoir. The assessment of viral production in cells from HIV-infected individuals with optimal viral suppression revealed the failure of SAHA/vorinostat to efficiently generate viral particle production. To further investigate and characterize the process of latency at the transcriptome level, and the response to SAHA as well as various reactivating agents, we use a model of primary CD4+ lymphocytes. The main observation from this study is that viral transcripts persist during latency, and that the accumulation of viral transcripts does not result in efficient viral protein expression upon reactivation with agents such as SAHA. Our data suggest that post-transcriptional blocks also contribute to latency, and that additional strategies need to be explored to efficiently purge the viral reservoir.
Collapse
Affiliation(s)
- Pejman Mohammadi
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel and Lausanne, Switzerland
| | - Julia di Iulio
- Swiss Institute of Bioinformatics, Basel and Lausanne, Switzerland
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Miguel Muñoz
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Raquel Martinez
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - István Bartha
- Swiss Institute of Bioinformatics, Basel and Lausanne, Switzerland
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Christian Thorball
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Jacques Fellay
- Swiss Institute of Bioinformatics, Basel and Lausanne, Switzerland
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel and Lausanne, Switzerland
- * E-mail: (NB); (AC); (AT)
| | - Angela Ciuffi
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
- * E-mail: (NB); (AC); (AT)
| | - Amalio Telenti
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
- * E-mail: (NB); (AC); (AT)
| |
Collapse
|
328
|
Dual role of novel ingenol derivatives from Euphorbia tirucalli in HIV replication: inhibition of de novo infection and activation of viral LTR. PLoS One 2014; 9:e97257. [PMID: 24827152 PMCID: PMC4020785 DOI: 10.1371/journal.pone.0097257] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/16/2014] [Indexed: 11/19/2022] Open
Abstract
HIV infection is not cleared by antiretroviral drugs due to the presence of latently infected cells that are not eliminated with current therapies and persist in the blood and organs of infected patients. New compounds to activate these latent reservoirs have been evaluated so that, along with HAART, they can be used to activate latent virus and eliminate the latently infected cells resulting in eradication of viral infection. Here we describe three novel diterpenes isolated from the sap of Euphorbia tirucalli, a tropical shrub. These molecules, identified as ingenols, were modified at carbon 3 and termed ingenol synthetic derivatives (ISD). They activated the HIV-LTR in reporter cell lines and human PBMCs with latent virus in concentrations as low as 10 nM. ISDs were also able to inhibit the replication of HIV-1 subtype B and C in MT-4 cells and human PBMCs at concentrations of EC50 0.02 and 0.09 µM respectively, which are comparable to the EC50 of some antiretroviral currently used in AIDS treatment. Control of viral replication may be caused by downregulation of surface CD4, CCR5 and CXCR4 observed after ISD treatment in vitro. These compounds appear to be less cytotoxic than other diterpenes such as PMA and prostratin, with effective dose versus toxic dose TI>400. Although the mechanisms of action of the three ISDs are primarily attributed to the PKC pathway, downregulation of surface receptors and stimulation of the viral LTR might be differentially modulated by different PKC isoforms.
Collapse
|
329
|
Measuring reversal of HIV-1 latency ex vivo using cells from infected individuals. Proc Natl Acad Sci U S A 2014; 111:6860-1. [PMID: 24799684 DOI: 10.1073/pnas.1405194111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
330
|
Badley AD. "Much ado to achieve nothing: prospects for curing HIV infection". MOLECULAR AND CELLULAR THERAPIES 2014; 2:9. [PMID: 26056578 PMCID: PMC4452067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/12/2014] [Indexed: 11/21/2023]
Abstract
Currently there is significant scientific effort being directed at developing ways to create either a sterilizing cure, or functional cure for HIV infection. Multiple approaches are being evaluated under the broad headings of gene therapy, immune based interventions, and treatments which depend upon HIV reactivation from latency to cause the death of cells which harbor the virus. Molecular and Cellular Therapies (MCT) welcomes all manuscripts devoted to increasing our understanding of determinants of affecting a cure for HIV and mechanistic studies determine the cellular and viral interventions necessary for achieving HIV cure.
Collapse
Affiliation(s)
- Andrew D Badley
- />Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota USA
- />Division of Molecular Medicine, Mayo Clinic, Rochester, Minnesota USA
| |
Collapse
|
331
|
Badley AD. "Much ado to achieve nothing: prospects for curing HIV infection". MOLECULAR AND CELLULAR THERAPIES 2014; 2:9. [PMID: 26056578 PMCID: PMC4452067 DOI: 10.1186/2052-8426-2-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/12/2014] [Indexed: 11/10/2022]
Abstract
Currently there is significant scientific effort being directed at developing ways to create either a sterilizing cure, or functional cure for HIV infection. Multiple approaches are being evaluated under the broad headings of gene therapy, immune based interventions, and treatments which depend upon HIV reactivation from latency to cause the death of cells which harbor the virus. Molecular and Cellular Therapies (MCT) welcomes all manuscripts devoted to increasing our understanding of determinants of affecting a cure for HIV and mechanistic studies determine the cellular and viral interventions necessary for achieving HIV cure.
Collapse
Affiliation(s)
- Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota USA ; Division of Molecular Medicine, Mayo Clinic, Rochester, Minnesota USA
| |
Collapse
|
332
|
Chang CC, Lewin SR. Challenges, progress and strategies in the search for a cure for HIV. MICROBIOLOGY AUSTRALIA 2014. [DOI: 10.1071/ma14023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|