51
|
Cellular Determinants of HIV Persistence on Antiretroviral Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1075:213-239. [PMID: 30030795 DOI: 10.1007/978-981-13-0484-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The era of antiretroviral therapy has made HIV-1 infection a manageable chronic disease for those with access to treatment. Despite treatment, virus persists in tissue reservoirs seeded with long-lived infected cells that are resistant to cell death and immune recognition. Which cells contribute to this reservoir and which factors determine their persistence are central questions that need to be answered to achieve viral eradication. In this chapter, we describe how cell susceptibility to infection, resistance to cell death, and immune-mediated killing as well as natural cell life span and turnover potential are central components that allow persistence of different lymphoid and myeloid cell subsets that were recently identified as key players in harboring latent and actively replicating virus. The relative contribution of these subsets to persistence of viral reservoir is described, and the open questions are highlighted.
Collapse
|
52
|
Siddiqui S, Perez S, Gao Y, Doyle-Meyers L, Foley BT, Li Q, Ling B. Persistent Viral Reservoirs in Lymphoid Tissues in SIV-Infected Rhesus Macaques of Chinese-Origin on Suppressive Antiretroviral Therapy. Viruses 2019; 11:v11020105. [PMID: 30691203 PMCID: PMC6410399 DOI: 10.3390/v11020105] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
Understanding HIV latent reservoirs in tissues is essential for the development of new strategies targeting these sites for eradication. Here, we assessed the size of latent reservoirs and the source of residual viruses in multiple lymphoid tissues of SIV-infected and fully suppressed rhesus macaques of Chinese-origin (cRMs). Eight cRMs were infected with SIVmac251 and treated with tenofovir and emtricitabine daily for 24 weeks initiated 4 weeks post-infection. Four of the eight animals reached sustained full viral suppression with undetectable viremia. The levels of cell-associated SIV DNA varied in peripheral blood mononuclear cells (PBMCs) and multiple lymphoid tissues, but with higher levels in the mesenteric lymph nodes (MesLNs). The levels of cell-associated SIV RNA also varied in different tissues. The higher frequency of viral RNA detection in the MesLNs was also observed by in situ hybridization. Consistently, the infection unit per million cells (IUPM) in the MesLNs was higher than in PBMCs and other tested lymphoid tissues by quantitative viral outgrowth assay (QVOA). Furthermore, env gp120 from tissue SIV RNA was amplified by single genome amplification. Phylogenetic analysis revealed diverse variants from tissues parallel to the viral inoculum in all viral suppressed animals. These results demonstrate that the latency and viral reservoirs in the lymphoid tissues still exist in aviremic macaques under full suppressive therapy. Moreover, the size of viral latent reservoirs differs in various lymphoid tissues with a relatively larger size in the MesLNs.
Collapse
Affiliation(s)
- Summer Siddiqui
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA.
| | - Stefanie Perez
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA.
| | - Yong Gao
- Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Lara Doyle-Meyers
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA 70433, USA.
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Brian T Foley
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Binhua Ling
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA.
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA.
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
53
|
Cao J, Zhang C, Han X, Cheng H, Chen W, Qi K, Qiao J, Sun Z, Wu Q, Zeng L, Niu M, Li L, Xu K. Emerging role of stem cell memory-like T cell in immune thrombocytopenia. Scand J Immunol 2019; 89:e12739. [PMID: 30506564 DOI: 10.1111/sji.12739] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Jiang Cao
- Department of Hematology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
| | - Changxiao Zhang
- Department of Endocrinology; Xuzhou City Hospital of Traditional Chinese Medicine; Xuzhou China
| | - Xiao Han
- Department of Hematology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
| | - Hai Cheng
- Department of Hematology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
| | - Wei Chen
- Department of Hematology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
| | - Kunming Qi
- Department of Hematology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
| | - Jianlin Qiao
- Jiangsu Bone Marrow Stem Cell Institute; Xuzhou China
| | - Zengtian Sun
- Jiangsu Bone Marrow Stem Cell Institute; Xuzhou China
| | - Qingyun Wu
- Jiangsu Bone Marrow Stem Cell Institute; Xuzhou China
| | - Lingyu Zeng
- Jiangsu Bone Marrow Stem Cell Institute; Xuzhou China
| | - Mingshan Niu
- Jiangsu Bone Marrow Stem Cell Institute; Xuzhou China
| | - Li Li
- Department of Gastroenterology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
| | - Kailin Xu
- Department of Hematology; The Affiliated Hospital of Xuzhou Medical University; Xuzhou China
- Jiangsu Bone Marrow Stem Cell Institute; Xuzhou China
| |
Collapse
|
54
|
Psomas CK. Of HIV and men. J Virus Erad 2019. [DOI: 10.1016/s2055-6640(20)30272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
55
|
Psomas CK. Of HIV and men. J Virus Erad 2019; 5:1-2. [PMID: 30800419 PMCID: PMC6362903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
56
|
Probabilistic control of HIV latency and transactivation by the Tat gene circuit. Proc Natl Acad Sci U S A 2018; 115:12453-12458. [PMID: 30455316 DOI: 10.1073/pnas.1811195115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The reservoir of HIV latently infected cells is the major obstacle for eradication of HIV infection. The "shock-and-kill" strategy proposed earlier aims to reduce the reservoir by activating cells out of latency. While the intracellular HIV Tat gene circuit is known to play important roles in controlling latency and its transactivation in HIV-infected cells, the detailed control mechanisms are not well understood. Here we study the mechanism of probabilistic control of the latent and the transactivated cell phenotypes of HIV-infected cells. We reconstructed the probability landscape, which is the probability distribution of the Tat gene circuit states, by directly computing the exact solution of the underlying chemical master equation. Results show that the Tat circuit exhibits a clear bimodal probability landscape (i.e., there are two distinct probability peaks, one associated with the latent cell phenotype and the other with the transactivated cell phenotype). We explore potential modifications to reactions in the Tat gene circuit for more effective transactivation of latent cells (i.e., the shock-and-kill strategy). Our results suggest that enhancing Tat acetylation can dramatically increase Tat and viral production, while increasing the Tat-transactivation response binding affinity can transactivate latent cells more rapidly than other manipulations. Our results further explored the "block and lock" strategy toward a functional cure for HIV. Overall, our study demonstrates a general approach toward discovery of effective therapeutic strategies and druggable targets by examining control mechanisms of cell phenotype switching via exactly computed probability landscapes of reaction networks.
Collapse
|
57
|
Tomalka AG, Resto-Garay I, Campbell KS, Popkin DL. In vitro Evidence That Combination Therapy With CD16-Bearing NK-92 Cells and FDA-Approved Alefacept Can Selectively Target the Latent HIV Reservoir in CD4+ CD2hi Memory T Cells. Front Immunol 2018; 9:2552. [PMID: 30455699 PMCID: PMC6230627 DOI: 10.3389/fimmu.2018.02552] [Citation(s) in RCA: 4] [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: 06/18/2018] [Accepted: 10/17/2018] [Indexed: 12/24/2022] Open
Abstract
Elimination of the latent HIV reservoir remains the biggest hurdle to achieve HIV cure. In order to specifically eliminate HIV infected cells they must be distinguishable from uninfected cells. CD2 was recently identified as a potential marker enriched in the HIV-1 reservoir on CD4+ T cells, the largest, longest-lived and best-characterized constituent of the HIV reservoir. We previously proposed to repurpose FDA-approved alefacept, a humanized α-CD2 fusion protein, to reduce the HIV reservoir in CD2hi CD4+ memory T cells. Here, we show the first evidence that alefacept can specifically target and reduce CD2hi HIV infected cells in vitro. We explore a variety of natural killer (NK) cells as mediators of antibody-dependent cell-mediated cytotoxicity (ADCC) including primary NK cells, expanded NK cells as well as the CD16 transduced NK-92 cell line which is currently under study in clinical trials as a treatment for cancer. We demonstrate that CD16.NK-92 has a natural preference to kill CD2hi CD45RA- memory T cells, specifically CD45RA- CD27+ central memory/transitional memory (TCM/TM) subset in both healthy and HIV+ patient samples as well as to reduce HIV DNA from HIV+ samples from donors well controlled on antiretroviral therapy. Lastly, alefacept can combine with CD16.NK-92 to decrease HIV DNA in some patient samples and thus may yield value as part of a strategy toward sustained HIV remission.
Collapse
Affiliation(s)
- Amanda G. Tomalka
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Ivelisse Resto-Garay
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Kerry S. Campbell
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Institute for Cancer Research, Philadelphia, PA, United States
| | - Daniel L. Popkin
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
58
|
Antibody-Mediated CD4 Depletion Induces Homeostatic CD4 + T Cell Proliferation without Detectable Virus Reactivation in Antiretroviral Therapy-Treated Simian Immunodeficiency Virus-Infected Macaques. J Virol 2018; 92:JVI.01235-18. [PMID: 30185596 DOI: 10.1128/jvi.01235-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
A major barrier to human immunodeficiency virus (HIV) eradication is the long-term persistence of latently infected CD4+ T cells harboring integrated replication-competent virus. It has been proposed that the homeostatic proliferation of these cells drives long-term reservoir persistence in the absence of virus reactivation, thus avoiding cell death due to either virus-mediated cytopathicity or immune effector mechanisms. Here, we conducted an experimental depletion of CD4+ T cells in eight antiretroviral therapy (ART)-treated, simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs) to determine whether the homeostatically driven CD4+ T-cell proliferation that follows CD4+ T-cell depletion results in reactivation of latent virus and/or expansion of the virus reservoir. After administration of the CD4R1 antibody, we observed a CD4+ T cell depletion of 65 to 89% in peripheral blood and 20 to 50% in lymph nodes, followed by a significant increase in CD4+ T cell proliferation during CD4+ T cell reconstitution. However, this CD4+ T cell proliferation was not associated with detectable increases in viremia, indicating that the homeostatic activation of CD4+ T cells is not sufficient to induce virus reactivation from latently infected cells. Interestingly, the homeostatic reconstitution of the CD4+ T cell pool was not associated with significant changes in the number of circulating cells harboring SIV DNA compared to results for the first postdepletion time point. This study indicates that, in ART-treated SIV-infected RMs, the homeostasis-driven CD4+ T-cell proliferation that follows experimental CD4+ T-cell depletion occurs in the absence of detectable reactivation of latent virus and does not increase the size of the virus reservoir as measured in circulating cells.IMPORTANCE Despite successful suppression of HIV replication with antiretroviral therapy, current treatments are unable to eradicate the latent virus reservoir, and treatment interruption almost invariably results in the reactivation of HIV even after decades of virus suppression. Homeostatic proliferation of latently infected cells is one mechanism that could maintain the latent reservoir. To understand the impact of homeostatic mechanisms on virus reactivation and reservoir size, we experimentally depleted CD4+ T cells in ART-treated SIV-infected rhesus macaques and monitored their homeostatic rebound. We find that depletion-induced proliferation of CD4+ T cells is insufficient to reactivate the viral reservoir in vivo Furthermore, the proportion of SIV DNA+ CD4+ T cells remains unchanged during reconstitution, suggesting that the reservoir is resistant to this mechanism of expansion at least in this experimental system. Understanding how T cell homeostasis impacts latent reservoir longevity could lead to the development of new treatment paradigms aimed at curing HIV infection.
Collapse
|
59
|
Rocco J, Mellors JW, Macatangay BJC. Regulatory T cells: the ultimate HIV reservoir? J Virus Erad 2018; 4:209-214. [PMID: 30515299 PMCID: PMC6248834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Despite significant advances in the understanding of HIV-1 infection, a cure remains out of reach. This is, in part, due to a long-lived HIV-1 reservoir in resting CD4+ T cells, which do not express viral antigens, and thus are invisible to the immune system. These latently infected cells carry replication-competent proviruses and can cause rebound viraemia if antiretrovirals are interrupted. Characterising this HIV-1 reservoir is a challenging task, requiring identification of CD4+ T cell subsets carrying intact proviruses, as well as defining their distribution within the body. Regulatory T cells (Tregs) comprise a subset of CD4+ T cells that are essential for maintaining immune tolerance. HIV-1 is known to infect Tregs in vivo but there is limited understanding of their role in HIV-1 persistence. Recent studies of well-controlled HIV-1 infection on antiretroviral therapy (ART) have shown higher frequencies of inducible, intact proviruses in Tregs compared to other CD4+ T cells, and provirus-containing Tregs have been found in lymphoid tissues at substantial frequencies. This evidence is supportive of a latent HIV-1 reservoir in Tregs, but greater detail is needed, including tissue distribution. An HIV-1 reservoir in Tregs could pose a significant barrier to HIV-1 eradication because Tregs are known to be long lived and resistant to apoptosis. Tregs are also immunosuppressive, and can inhibit cell-mediated immunity through multiple mechanisms. Non-specific depletion of Tregs would be likely to result in severe autoimmunity. Additional research is needed to further characterise regulatory T cells as a reservoir of HIV-1 and as an obstacle to eradication, or immune control, of HIV-1 infection.
Collapse
Affiliation(s)
| | | | - Bernard JC Macatangay
- Corresponding author: Bernard JC Macatangay,
S827 Scaife Hall, 3550 Terrace Street,
Pittsburgh,
PA15261,
USA
| |
Collapse
|
60
|
|
61
|
Abstract
HIV integrates into the host genome to create a persistent viral reservoir. Stimulation of CD4+ memory T lymphocytes with common γc-chain cytokines renders these cells more susceptible to HIV infection, making them a key component of the reservoir itself. IL-15 is up-regulated during primary HIV infection, a time when the HIV reservoir established. Therefore, we investigated the molecular and cellular impact of IL-15 on CD4+ T-cell infection. We found that IL-15 stimulation induces SAM domain and HD domain-containing protein 1 (SAMHD1) phosphorylation due to cell cycle entry, relieving an early block to infection. Perturbation of the pathways downstream of IL-15 receptor (IL-15R) indicated that SAMHD1 phosphorylation after IL-15 stimulation is JAK dependent. Treating CD4+ T cells with Ruxolitinib, an inhibitor of JAK1 and JAK2, effectively blocked IL-15-induced SAMHD1 phosphorylation and protected CD4+ T cells from HIV infection. Using high-resolution single-cell immune profiling using mass cytometry by TOF (CyTOF), we found that IL-15 stimulation altered the composition of CD4+ T-cell memory populations by increasing proliferation of memory CD4+ T cells, including CD4+ T memory stem cells (TSCM). IL-15-stimulated CD4+ TSCM, harboring phosphorylated SAMHD1, were preferentially infected. We propose that IL-15 plays a pivotal role in creating a self-renewing, persistent HIV reservoir by facilitating infection of CD4+ T cells with stem cell-like properties. Time-limited interventions with JAK1 inhibitors, such as Ruxolitinib, should prevent the inactivation of the endogenous restriction factor SAMHD1 and protect this long-lived CD4+ T-memory cell population from HIV infection.
Collapse
|
62
|
Anderson EM, Maldarelli F. Quantification of HIV DNA Using Droplet Digital PCR Techniques. ACTA ACUST UNITED AC 2018; 51:e62. [PMID: 30253074 DOI: 10.1002/cpmc.62] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
HIV persists, despite effective antiretroviral therapy, in long-lived cells, posing a major barrier toward a cure. A key step in the HIV replication cycle and a hallmark of the Retroviridae family is the integration of the viral DNA into the host genome. Once integrated, HIV expression is regulated by host machinery and the provirus persists until the cell dies. A reservoir of cells harboring replication-competent proviruses can survive for years, and mechanisms that maintain that reservoir are under investigation. The majority of integrated proviruses, however, are defective or have large deletions, and the composition of the proviral landscape during therapy remains unknown. Methods to quantify HIV proviruses are useful in investigating HIV persistence. Presented in this unit is a method for total HIV DNA quantification of various HIV genome targets that utilizes the next-generation PCR platform, digital PCR. The abundance of various HIV gene targets reflects the overall proviral composition. In this protocol, total genomic DNA is isolated from patient-derived cells and then used as a template for droplet digital PCR, in which the PCR reaction is partitioned into approximately 20,000 individual droplets, PCR amplified to an end point, and subjected to absolute quantification by counting the number of positive and negative droplets. Copy number is directly calculated using straightforward Poisson correction. Additionally, this methodological approach can be used to obtain absolute quantification of other DNA targets. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, NCI, NIH, Frederick, Maryland
| |
Collapse
|
63
|
Kumar NA, van der Sluis RM, Mota T, Pascoe R, Evans VA, Lewin SR, Cameron PU. Myeloid Dendritic Cells Induce HIV Latency in Proliferating CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1468-1477. [PMID: 30030324 DOI: 10.4049/jimmunol.1701233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 06/27/2018] [Indexed: 02/04/2023]
Abstract
HIV latency occurs predominantly in long-lived resting CD4+ T cells; however, latent infection also occurs in T cell subsets, including proliferating CD4+ T cells. We compared the establishment and maintenance of latent infection in nonproliferating and proliferating human CD4+ T cells cocultured with syngeneic myeloid dendritic cells (mDC). Resting CD4+ T cells were labeled with the proliferation dye eFluor 670 and cultured alone or with mDC, plasmacytoid dendritic cells, or monocytes in the presence of staphylococcal enterotoxin B (SEB). Cells were cultured for 24 h and infected with CCR5-tropic enhanced GFP (EGFP) reporter HIV. Five days postinfection, nonproductively infected EGFP- CD4+ T cells that were either nonproliferating (eFluor 670hi) or proliferating (eFluor 670lo) were sorted and cultured for an additional 7 d (day 12) with IL-7 and antiretrovirals. At day 5 postinfection, sorted, nonproductively infected T cells were stimulated with anti-CD3/CD28, and induced expression of EGFP was measured to determine the frequency of latent infection. Integrated HIV in these cells was confirmed using quantitative PCR. By these criteria, latent infection was detected at day 5 and 12 in proliferating T cells cocultured with mDC and monocytes but not plasmacytoid dendritic cells, where CD4+ T cells at day 12 were poor. At day 5 postinfection, nonproliferating T cells expressing SEB-specific TCR Vβ-17 were enriched in latent infection compared with non-SEB-specific TCR Vβ-8.1. Together, these data show that both nonproliferating and proliferating CD4+ T cells can harbor latent infection during SEB-stimulated T cell proliferation and that the establishment of HIV latency in nonproliferating T cells is linked to expression of specific TCR that respond to SEB.
Collapse
Affiliation(s)
- Nitasha A Kumar
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Renee M van der Sluis
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Talia Mota
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Rachel Pascoe
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Vanessa A Evans
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia; and.,Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Paul U Cameron
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia; .,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia; and.,Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| |
Collapse
|
64
|
Roychoudhury P, De Silva Feelixge H, Reeves D, Mayer BT, Stone D, Schiffer JT, Jerome KR. Viral diversity is an obligate consideration in CRISPR/Cas9 designs for targeting the HIV reservoir. BMC Biol 2018; 16:75. [PMID: 29996827 PMCID: PMC6040082 DOI: 10.1186/s12915-018-0544-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND RNA-guided CRISPR/Cas9 systems can be designed to mutate or excise the integrated HIV genome from latently infected cells and have therefore been proposed as a curative approach for HIV. However, most studies to date have focused on molecular clones with ideal target site recognition and do not account for target site variability observed within and between patients. For clinical success and broad applicability, guide RNA (gRNA) selection must account for circulating strain diversity and incorporate the within-host diversity of HIV. RESULTS We identified a set of gRNAs targeting HIV LTR, gag, and pol using publicly available sequences for these genes and ranked gRNAs according to global conservation across HIV-1 group M and within subtypes A-C. By considering paired and triplet combinations of gRNAs, we found triplet sets of target sites such that at least one of the gRNAs in the set was present in over 98% of all globally available sequences. We then selected 59 gRNAs from our list of highly conserved LTR target sites and evaluated in vitro activity using a loss-of-function LTR-GFP fusion reporter. We achieved efficient GFP knockdown with multiple gRNAs and found clustering of highly active gRNA target sites near the middle of the LTR. Using published deep-sequence data from HIV-infected patients, we found that globally conserved sites also had greater within-host target conservation. Lastly, we developed a mathematical model based on varying distributions of within-host HIV sequence diversity and enzyme efficacy. We used the model to estimate the number of doses required to deplete the latent reservoir and achieve functional cure thresholds. Our modeling results highlight the importance of within-host target site conservation. While increased doses may overcome low target cleavage efficiency, inadequate targeting of rare strains is predicted to lead to rebound upon cART cessation even with many doses. CONCLUSIONS Target site selection must account for global and within host viral genetic diversity. Globally conserved target sites are good starting points for design, but multiplexing is essential for depleting quasispecies and preventing viral load rebound upon therapy cessation.
Collapse
Affiliation(s)
| | | | - Daniel Reeves
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Daniel Stone
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, USA
- Department of Medicine, University of Washington, Seattle, USA
| | - Keith R Jerome
- Department of Laboratory Medicine, University of Washington, Seattle, USA.
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, USA.
| |
Collapse
|
65
|
Salantes DB, Zheng Y, Mampe F, Srivastava T, Beg S, Lai J, Li JZ, Tressler RL, Koup RA, Hoxie J, Abdel-Mohsen M, Sherrill-Mix S, McCormick K, Overton ET, Bushman FD, Learn GH, Siliciano RF, Siliciano JM, Tebas P, Bar KJ. HIV-1 latent reservoir size and diversity are stable following brief treatment interruption. J Clin Invest 2018; 128:3102-3115. [PMID: 29911997 PMCID: PMC6026010 DOI: 10.1172/jci120194] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/24/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The effect of a brief analytical treatment interruption (ATI) on the HIV-1 latent reservoir of individuals who initiate antiretroviral therapy (ART) during chronic infection is unknown. METHODS We evaluated the impact of transient viremia on the latent reservoir in participants who underwent an ATI and at least 6 months of subsequent viral suppression in a clinical trial testing the effect of passive infusion of the broadly neutralizing Ab VRC01 during ATI. RESULTS Measures of total HIV-1 DNA, cell-associated RNA, and infectious units per million cells (IUPM) (measured by quantitative viral outgrowth assay [QVOA]) were not statistically different before or after ATI. Phylogenetic analyses of HIV-1 env sequences from QVOA and proviral DNA demonstrated little change in the composition of the virus populations comprising the pre- and post-ATI reservoir. Expanded clones were common in both QVOA and proviral DNA sequences. The frequency of clonal populations differed significantly between QVOA viruses, proviral DNA sequences, and the viruses that reactivated in vivo. CONCLUSIONS The results indicate that transient viremia from ATI does not substantially alter measures of the latent reservoir, that clonal expansion is prevalent within the latent reservoir, and that characterization of latent viruses that can reactivate in vivo remains challenging. TRIAL REGISTRATION ClinicalTrials.gov NCT02463227FUNDING. Funding was provided by the NIH.
Collapse
Affiliation(s)
| | - Yu Zheng
- Harvard University, Cambridge, Massachusetts, USA
| | - Felicity Mampe
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Subul Beg
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jun Lai
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Randall L. Tressler
- Division of AIDS (DAIDS), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | | | - James Hoxie
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | - Robert F. Siliciano
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Pablo Tebas
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
66
|
Kuo HH, Ahmad R, Lee GQ, Gao C, Chen HR, Ouyang Z, Szucs MJ, Kim D, Tsibris A, Chun TW, Battivelli E, Verdin E, Rosenberg ES, Carr SA, Yu XG, Lichterfeld M. Anti-apoptotic Protein BIRC5 Maintains Survival of HIV-1-Infected CD4 + T Cells. Immunity 2018; 48:1183-1194.e5. [PMID: 29802019 DOI: 10.1016/j.immuni.2018.04.004] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/17/2018] [Accepted: 04/02/2018] [Indexed: 01/16/2023]
Abstract
HIV-1 infection of CD4+ T cells leads to cytopathic effects and cell demise, which is counter to the observation that certain HIV-1-infected cells possess a remarkable long-term stability and can persist lifelong in infected individuals treated with suppressive antiretroviral therapy (ART). Using quantitative mass spectrometry-based proteomics, we showed that HIV-1 infection activated cellular survival programs that were governed by BIRC5, a molecular inhibitor of cell apoptosis that is frequently overexpressed in malignant cells. BIRC5 and its upstream regulator OX40 were upregulated in productively and latently infected CD4+ T cells and were functionally involved in maintaining their viability. Moreover, OX40-expressing CD4+ T cells from ART-treated patients were enriched for clonally expanded HIV-1 sequences, and pharmacological inhibition of BIRC5 resulted in a selective decrease of HIV-1-infected cells in vitro. Together, these findings suggest that BIRC5 supports long-term survival of HIV-1-infected cells and may lead to clinical strategies to reduce persisting viral reservoirs.
Collapse
Affiliation(s)
- Hsiao-Hsuan Kuo
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rushdy Ahmad
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Guinevere Q Lee
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Hsiao-Rong Chen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zhengyu Ouyang
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Matthew J Szucs
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Dhohyung Kim
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Athe Tsibris
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Tae-Wook Chun
- National Institute of Allergies and Infectious Diseases, Bethesda, MD 20892, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Eric S Rosenberg
- Infectious Disease Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
67
|
García M, Buzón MJ, Benito JM, Rallón N. Peering into the HIV reservoir. Rev Med Virol 2018; 28:e1981. [PMID: 29744964 DOI: 10.1002/rmv.1981] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
Abstract
The main obstacle to HIV eradication is the establishment of a long-term persistent HIV reservoir. Although several therapeutic approaches have been developed to reduce and eventually eliminate the HIV reservoir, only a few have achieved promising results. A better knowledge of the mechanisms involved in the establishment and maintenance of HIV reservoir is of utmost relevance for the design of new therapeutic strategies aimed at purging it with the ultimate goal of achieving HIV eradication or alternatively a functional cure. In this regard, it is also important to take a close look into the cellular HIV reservoirs other than resting memory CD4 T-cells with key roles in reservoir maintenance that have been recently described. Unraveling the special characteristics of these HIV cellular compartments could aid us in designing new therapeutic strategies to deplete the latent HIV reservoir.
Collapse
Affiliation(s)
- Marcial García
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | | | - José M Benito
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Norma Rallón
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain.,Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| |
Collapse
|
68
|
Martin GE, Pace M, Thornhill JP, Phetsouphanh C, Meyerowitz J, Gossez M, Brown H, Olejniczak N, Lwanga J, Ramjee G, Kaleebu P, Porter K, Willberg CB, Klenerman P, Nwokolo N, Fox J, Fidler S, Frater J. CD32-Expressing CD4 T Cells Are Phenotypically Diverse and Can Contain Proviral HIV DNA. Front Immunol 2018; 9:928. [PMID: 29780387 PMCID: PMC5946760 DOI: 10.3389/fimmu.2018.00928] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/16/2018] [Indexed: 12/30/2022] Open
Abstract
Efforts to both characterize and eradicate the HIV reservoir have been limited by the rarity of latently infected cells and the absence of a specific denoting biomarker. CD32a (FcγRIIa) has been proposed to be a marker for an enriched CD4 T cell HIV reservoir, but this finding remains controversial. Here, we explore the expression of CD32 on CD3+CD4+ cells in participants from two primary HIV infection studies and identify at least three distinct phenotypes (CD32low, CD32+CD14+, and CD32high). Of note, CD4 negative enrichment kits remove the majority of CD4+CD32+ T cells, potentially skewing subsequent analyses if used. CD32high CD4 T cells had higher levels of HLA-DR and HIV co-receptor expression than other subsets, compatible with their being more susceptible to infection. Surprisingly, they also expressed high levels of CD20, TCRαβ, IgD, and IgM (but not IgG), markers for both T cells and naïve B cells. Compared with other populations, CD32low cells had a more differentiated memory phenotype and high levels of immune checkpoint receptors, programmed death receptor-1 (PD-1), Tim-3, and TIGIT. Within all three CD3+CD4+CD32+ phenotypes, cells could be identified in infected participants, which contained HIV DNA. CD32 expression on CD4 T cells did not correlate with HIV DNA or cell-associated HIV RNA (both surrogate measures of overall reservoir size) or predict time to rebound viremia following treatment interruption, suggesting that it is not a dominant biomarker for HIV persistence. Our data suggest that while CD32+ T cells can be infected with HIV, CD32 is not a specific marker of the reservoir although it might identify a population of HIV enriched cells in certain situations.
Collapse
Affiliation(s)
- Genevieve E Martin
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthew Pace
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - John P Thornhill
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Division of Medicine, Wright Fleming Institute, Imperial College, London, United Kingdom
| | - Chansavath Phetsouphanh
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jodi Meyerowitz
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Morgane Gossez
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Helen Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Natalia Olejniczak
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julianne Lwanga
- Department of Genitourinary Medicine and Infectious Disease, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Gita Ramjee
- HIV Prevention Research Unit, South African Medical Research Council, Durban, South Africa
| | | | - Kholoud Porter
- Research Department of Infection and Population Health, University College London, London, United Kingdom
| | - Christian B Willberg
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Nneka Nwokolo
- Chelsea and Westminster Hospital, London, United Kingdom
| | - Julie Fox
- Department of Genitourinary Medicine and Infectious Disease, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Sarah Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
69
|
McBrien JB, Kumar NA, Silvestri G. Mechanisms of CD8 + T cell-mediated suppression of HIV/SIV replication. Eur J Immunol 2018; 48:898-914. [PMID: 29427516 DOI: 10.1002/eji.201747172] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
In this article, we summarize the role of CD8+ T cells during natural and antiretroviral therapy (ART)-treated HIV and SIV infections, discuss the mechanisms responsible for their suppressive activity, and review the rationale for CD8+ T cell-based HIV cure strategies. Evidence suggests that CD8+ T cells are involved in the control of virus replication during HIV and SIV infections. During early HIV infection, the cytolytic activity of CD8+ T cells is responsible for control of viremia. However, it has been proposed that CD8+ T cells also use non-cytolytic mechanisms to control SIV infection. More recently, CD8+ T cells were shown to be required to fully suppress virus production in ART-treated SIV-infected macaques, suggesting that CD8+ T cells are involved in the control of virus transcription in latently infected cells that persist under ART. A better understanding of the complex antiviral activities of CD8+ T cells during HIV/SIV infection will pave the way for immune interventions aimed at harnessing these functions to target the HIV reservoir.
Collapse
Affiliation(s)
- Julia Bergild McBrien
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Nitasha A Kumar
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| |
Collapse
|
70
|
Pinzone MR, O’Doherty U. Measuring integrated HIV DNA ex vivo and in vitro provides insights about how reservoirs are formed and maintained. Retrovirology 2018; 15:22. [PMID: 29452580 PMCID: PMC5816390 DOI: 10.1186/s12977-018-0396-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/19/2018] [Indexed: 11/29/2022] Open
Abstract
The identification of the most appropriate marker to measure reservoir size has been a great challenge for the HIV field. Quantitative viral outgrowth assay (QVOA), the reference standard to quantify the amount of replication-competent virus, has several limitations, as it is laborious, expensive, and unable to robustly reactivate every single integrated provirus. PCR-based assays have been developed as an easier, cheaper and less error-prone alternative to QVOA, but also have limitations. Historically, measuring integrated HIV DNA has provided insights about how reservoirs are formed and maintained. In the 1990s, measuring integrated HIV DNA was instrumental in understanding that a subset of resting CD4 T cells containing integrated HIV DNA were the major source of replication-competent virus. Follow-up studies have further characterized the phenotype of these cells containing integrated HIV DNA, as well as shown the correlation between the integration levels and clinical parameters, such as duration of infection, CD4 count and viral load. Integrated HIV DNA correlates with total HIV measures and with QVOA. The integration assay has several limitations. First, it largely overestimates the reservoir size, as both defective and replication-competent proviruses are detected. Since defective proviruses are the majority in patients on ART, it follows that the number of proviruses capable of reactivating and releasing new virions is significantly smaller than the number of integrated proviruses. Second, in patients on ART clonal expansion could theoretically lead to the preferential amplification of proviruses close to an Alu sequence though longitudinal studies have not captured this effect. Proviral sequencing combined with integration measures is probably the best estimate of reservoir size, but it is expensive, time-consuming and requires considerable bioinformatics expertise. All these reasons limit its use on a large scale. Herein, we review the utility of measuring HIV integration and suggest combining it with sequencing and total HIV measurements can provide insights that underlie reservoir maintenance.
Collapse
Affiliation(s)
- Marilia Rita Pinzone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Una O’Doherty
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
71
|
Stein J, Storcksdieck Genannt Bonsmann M, Streeck H. Barriers to HIV Cure. HLA 2018; 88:155-63. [PMID: 27620852 DOI: 10.1111/tan.12867] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023]
Abstract
Since the beginning of the epidemic, more than 70 million people have been infected with human immunodeficiency virus (HIV) and about 38 million have died from acquired immune deficiency syndrome (AIDS)-related illnesses. While the discovery of highly active antiretroviral therapy (HAART) in the mid 90's has saved millions of lives, a complete eradication of HIV is still not possible as HIV can persist for decades in a small reservoir of latently infected cells. Once reactivated, these latently infected cells can actively produce viral particles. Recent studies suggest that several sanctuaries exist within infected individuals where HIV can remain undetected by the immune system. These cellular, anatomical and microanatomical viral reservoirs represent a major obstacle for the eradication of HIV. Here we review recent findings on potential sanctuaries of HIV and address potential avenues to overcome these immunological barriers.
Collapse
Affiliation(s)
- J Stein
- Institute for HIV Research, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | | | - H Streeck
- Institute for HIV Research, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,US Military HIV Research Program, Henry M. Jackson Foundation, Rockville, MD, USA
| |
Collapse
|
72
|
Modeling of Antilatency Treatment in HIV: What Is the Optimal Duration of Antiretroviral Therapy-Free HIV Remission? J Virol 2017; 91:JVI.01395-17. [PMID: 29021399 DOI: 10.1128/jvi.01395-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 10/03/2017] [Indexed: 11/20/2022] Open
Abstract
A number of treatment strategies are currently being developed to promote antiretroviral therapy-free HIV cure or remission. While complete elimination of the HIV reservoir would prevent recurrence of infection, it is not clear how different remission lengths would affect viral rebound and transmission. In this work, we use a stochastic model to show that a treatment that achieves a 1-year average time to viral remission will still lead to nearly a quarter of subjects experiencing viral rebound within the first 3 months. Given quarterly viral testing intervals, this leads to an expected 39 (95% uncertainty interval [UI], 22 to 69) heterosexual transmissions and up to 262 (95% UI, 107 to 534) homosexual transmissions per 1,000 treated subjects over a 10-year period. Thus, a balance between high initial treatment levels, risk of recrudescence, and risk of transmission should be considered when assessing the "useful" or optimal length of antiretroviral therapy-free HIV remission to be targeted. We also investigate the trade-off between increasing the average duration of remission versus the risk of treatment failure (viral recrudescence) and the need for retreatment. To minimize drug exposure, we found that the optimal target of antilatency interventions is a 1,700-fold reduction in the size of the reservoir, which leads to an average time to recrudescence of 30 years. Interestingly, this is a significantly lower level of reduction than that required for complete elimination of the viral reservoir. Additionally, we show that when shorter periods are targeted, there is a real probability of viral transmission occurring between tests for viral rebound.IMPORTANCE Current treatment of HIV involves patients taking antiretroviral therapy to ensure that the level of virus remains very low or undetectable. Continuous therapy is required, as the virus persists in a latent state within cells, and when therapy is stopped, the virus rebounds, usually within 2 weeks. A major question is how to reduce the amount of persistent virus and therefore allow a delay or remission until the virus returns after ceasing therapy. In this work, we consider the probability that HIV will still rebound even after this reduction and ask what the likelihood of viral transmission would be in this case.
Collapse
|
73
|
Khan S, Telwatte S, Trapecar M, Yukl S, Sanjabi S. Differentiating Immune Cell Targets in Gut-Associated Lymphoid Tissue for HIV Cure. AIDS Res Hum Retroviruses 2017; 33:S40-S58. [PMID: 28882067 PMCID: PMC5685216 DOI: 10.1089/aid.2017.0153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The single greatest challenge to an HIV cure is the persistence of latently infected cells containing inducible, replication-competent proviral genomes, which constitute only a small fraction of total or infected cells in the body. Although resting CD4+ T cells in the blood are a well-known source of viral rebound, more than 90% of the body's lymphocytes reside elsewhere. Many are in gut tissue, where HIV DNA levels per million CD4+ T cells are considerably higher than in the blood. Despite the significant contribution of gut tissue to viral replication and persistence, little is known about the cell types that support persistence of HIV in the gut; importantly, T cells in the gut have phenotypic, functional, and survival properties that are distinct from T cells in other tissues. The mechanisms by which latency is established and maintained will likely depend on the location and cytokine milieu surrounding the latently infected cells in each compartment. Therefore, successful HIV cure strategies require identification and characterization of the exact cell types that support viral persistence, particularly in the gut. In this review, we describe the seeding of the latent HIV reservoir in the gut mucosa; highlight the evidence for compartmentalization and depletion of T cells; summarize the immunologic consequences of HIV infection within the gut milieu; propose how the damaged gut environment may promote the latent HIV reservoir; and explore several immune cell targets in the gut and their place on the path toward HIV cure.
Collapse
Affiliation(s)
- Shahzada Khan
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
| | - Sushama Telwatte
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Martin Trapecar
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
| | - Steven Yukl
- San Francisco VA Health Care System and University of California, San Francisco (UCSF), San Francisco, California
| | - Shomyseh Sanjabi
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
74
|
Lu X, Song B, Weng W, Xia H, Su B, Wu H, Zhang T, Li W, Gao Y. CD4 + T Memory Stem Cells Correlate with Disease Progression in Chronically HIV-1-Infected Patients. Viral Immunol 2017; 30:642-648. [PMID: 29035156 DOI: 10.1089/vim.2017.0017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recently identified T memory stem (Tscm) cells have stem-cell-like properties, including long lifespan, self-renewal capacity, and multipotency to differentiate into other memory T cell types. In the study of simian immunodeficiency virus (SIV) infection, selective depletion of CCR5+CD4+ Tscm cells and the high proliferation rate of these cells are believed to be responsible for the pathogenesis of SIV-infected rhesus macaques. Here, we conducted a cohort study to investigate the influence of chronic human immunodeficiency virus (HIV)-1 infection on CD4+ Tscm cell homeostasis, and the effect of antiretroviral therapy (ART) on CD4+ Tscm cells. Chronic HIV-1 infection resulted in a decrease of the CD4+ Tscm cell proportion in HIV-1 patients. The decreased number of CD4+ Tscm cells in HIV-1 patients correlated positively with that of circulating CD4+ T cells. Further, the depletion of CD4+ Tscm cells was inversely correlated with an increased level of T cell immune activation during chronic HIV-1 infection. Prolonged ART recovered the CD4+ Tscm cells, and the dynamic change of CD4+ Tscm cells was in parallel with CD4+ T cell restoration and a decrease in the level of T cell immune activation. We propose that the abnormity of CD4+ Tscm cells may contribute to the pathogenesis and disease progression in HIV-1-infected individuals.
Collapse
Affiliation(s)
- Xiaofan Lu
- 1 Beijing Key Laboratory for HIV/AIDS Research , Beijing, People's Republic of China .,2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| | - Bingbing Song
- 2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| | - Wenjia Weng
- 2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| | - Huan Xia
- 1 Beijing Key Laboratory for HIV/AIDS Research , Beijing, People's Republic of China
| | - Bin Su
- 1 Beijing Key Laboratory for HIV/AIDS Research , Beijing, People's Republic of China .,2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| | - Hao Wu
- 2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| | - Tong Zhang
- 2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| | - Wei Li
- 3 Center for Interventional Oncology, Beijing You'an Hospital, Capital Medical University , Beijing, People's Republic of China
| | - Yanqing Gao
- 2 Center for Infectious Disease, Capital Medical University , Beijing, People's Republic of China
| |
Collapse
|
75
|
Hernandez-Vargas EA. Modeling Kick-Kill Strategies toward HIV Cure. Front Immunol 2017; 8:995. [PMID: 28894444 PMCID: PMC5581319 DOI: 10.3389/fimmu.2017.00995] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/04/2017] [Indexed: 12/31/2022] Open
Abstract
Although combinatorial antiretroviral therapy (cART) potently suppresses the virus, a sterile or functional cure still remains one of the greatest therapeutic challenges worldwide. Reservoirs are infected cells that can maintain HIV persistence for several years in patients with optimal cART, which is a leading obstacle to eradicate the virus. Despite the significant progress that has been made in our understanding of the diversity of cells that promote HIV persistence, many aspects that are critical to the development of effective therapeutic approaches able to purge the latent CD4+ T cell reservoir are poorly understood. Simultaneous purging strategies known as “kick-kill” have been pointed out as promising therapeutic approaches to eliminate the viral reservoir. However, long-term outcomes of purging strategies as well as the effect on the HIV reservoir are still largely fragmented. In this context, mathematical modeling can provide a rationale not only to evaluate the impact on the HIV reservoir but also to facilitate the formulation of hypotheses about potential therapeutic strategies. This review aims to discuss briefly the most recent mathematical modeling contributions, harnessing our knowledge toward the uncharted territory of HIV eradication. In addition, problems associated with current models are discussed, in particular, mathematical models consider only T cell responses but HIV control may also depend on other cell responses as well as chemokines and cytokines dynamics.
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW HIV-1 is able to create lasting reservoirs of virally infected cells that persist life-long and are extremely difficult to eradicate, thus necessitating indefinite antiretroviral therapy. Large numbers of studies suggest that CD4 T cells represent the major, and possibly the only cell type supporting HIV-1 long-term persistence. However, the ability to serve as long-term viral reservoirs may be confined to certain subpopulations of CD4 T cells with specific functional and developmental characteristics that HIV-1 can selectively exploit to propagate long-term viral survival within the host. Identification of CD4 T-cell subtypes that serve as hotspots for viral persistence may be critical for designing strategies to purge the immune system of persisting viral reservoirs. RECENT FINDINGS Developmentally immature, long-lasting CD4 memory T-cell populations seem to contain the majority of latently HIV-1-infected cells that persist despite antiretroviral therapy in the peripheral blood. Emerging data suggest that functional polarization toward a T helper 17 (Th17), a T follicular helper cell or a regulatory T-cell lineage may also be associated with an increased ability to serve as a viral reservoir site. Atypical T cells such a γδ CD4 T cells or tissue-resident memory CD4 T cells may be predestined to serve as sites for HIV-1 persistence in specific tissues, but will require additional exploration in future studies. SUMMARY Recent advances have increased awareness for the profound diversity and complexity of CD4 T-cell subpopulations serving as sites for HIV-1 persistence. Continuous technological and methodological improvements to interrogate viral reservoirs in distinct CD4 T-cell subpopulations may allow to define a more complete landscape of the HIV-1 reservoir composition in different T-cell subpopulations.
Collapse
|
77
|
Murray AJ, Kwon KJ, Farber DL, Siliciano RF. The Latent Reservoir for HIV-1: How Immunologic Memory and Clonal Expansion Contribute to HIV-1 Persistence. THE JOURNAL OF IMMUNOLOGY 2017; 197:407-17. [PMID: 27382129 DOI: 10.4049/jimmunol.1600343] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/12/2016] [Indexed: 12/15/2022]
Abstract
Combination antiretroviral therapy (ART) for HIV-1 infection reduces plasma virus levels to below the limit of detection of clinical assays. However, even with prolonged suppression of viral replication with ART, viremia rebounds rapidly after treatment interruption. Thus, ART is not curative. The principal barrier to cure is a remarkably stable reservoir of latent HIV-1 in resting memory CD4(+) T cells. In this review, we consider explanations for the remarkable stability of the latent reservoir. Stability does not appear to reflect replenishment from new infection events but rather normal physiologic processes that provide for immunologic memory. Of particular importance are proliferative processes that drive clonal expansion of infected cells. Recent evidence suggests that in some infected cells, proliferation is a consequence of proviral integration into host genes associated with cell growth. Efforts to cure HIV-1 infection by targeting the latent reservoir may need to consider the potential of latently infected cells to proliferate.
Collapse
Affiliation(s)
- Alexandra J Murray
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kyungyoon J Kwon
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032; Department of Surgery, Columbia University Medical Center, New York, NY 10032; and
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; Howard Hughes Medical Institute, Baltimore MD 21250
| |
Collapse
|
78
|
Gattinoni L, Speiser DE, Lichterfeld M, Bonini C. T memory stem cells in health and disease. Nat Med 2017; 23:18-27. [PMID: 28060797 DOI: 10.1038/nm.4241] [Citation(s) in RCA: 445] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/28/2016] [Indexed: 12/11/2022]
Abstract
T memory stem (TSCM) cells are a rare subset of memory lymphocytes endowed with the stem cell-like ability to self-renew and the multipotent capacity to reconstitute the entire spectrum of memory and effector T cell subsets. Cumulative evidence in mice, nonhuman primates and humans indicates that TSCM cells are minimally differentiated cells at the apex of the hierarchical system of memory T lymphocytes. Here we describe emerging findings demonstrating that TSCM cells, owing to their extreme longevity and robust potential for immune reconstitution, are central players in many physiological and pathological human processes. We also discuss how TSCM cell stemness could be leveraged therapeutically to enhance the efficacy of vaccines and adoptive T cell therapies for cancer and infectious diseases or, conversely, how it could be disrupted to treat TSCM cell driven and sustained diseases, such as autoimmunity, adult T cell leukemia and HIV-1.
Collapse
Affiliation(s)
- Luca Gattinoni
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel E Speiser
- Department of Oncology, Ludwig Cancer Research, Lausanne University Hospital, Lausanne, Switzerland
| | - Mathias Lichterfeld
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology Transplantation and Infectious Diseases, Leukemia Unit, San Raffaele Scientific Institute, Milan, Italy.,Hematology Department, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
79
|
Reeves DB, Duke ER, Hughes SM, Prlic M, Hladik F, Schiffer JT. Anti-proliferative therapy for HIV cure: a compound interest approach. Sci Rep 2017. [PMID: 28638104 PMCID: PMC5479830 DOI: 10.1038/s41598-017-04160-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In the era of antiretroviral therapy (ART), HIV-1 infection is no longer tantamount to early death. Yet the benefits of treatment are available only to those who can access, afford, and tolerate taking daily pills. True cure is challenged by HIV latency, the ability of chromosomally integrated virus to persist within memory CD4+ T cells in a non-replicative state and activate when ART is discontinued. Using a mathematical model of HIV dynamics, we demonstrate that treatment strategies offering modest but continual enhancement of reservoir clearance rates result in faster cure than abrupt, one-time reductions in reservoir size. We frame this concept in terms of compounding interest: small changes in interest rate drastically improve returns over time. On ART, latent cell proliferation rates are orders of magnitude larger than activation and new infection rates. Contingent on subtypes of cells that may make up the reservoir and their respective proliferation rates, our model predicts that coupling clinically available, anti-proliferative therapies with ART could result in functional cure within 2–10 years rather than several decades on ART alone.
Collapse
Affiliation(s)
- Daniel B Reeves
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, 98109, USA
| | - Elizabeth R Duke
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, 98109, USA.,University of Washington, Department of Medicine, Seattle, WA, 98195, USA
| | - Sean M Hughes
- University of Washington, Departments of Obstetrics and Gynecology, Seattle, WA, 98195, USA
| | - Martin Prlic
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, 98109, USA.,University of Washington, Department of Global Health, Seattle, WA, 98105, USA
| | - Florian Hladik
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, 98109, USA. .,University of Washington, Departments of Obstetrics and Gynecology, Seattle, WA, 98195, USA.
| | - Joshua T Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, 98109, USA. .,University of Washington, Department of Medicine, Seattle, WA, 98195, USA. .,Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, 98109, USA.
| |
Collapse
|
80
|
Lee GQ, Orlova-Fink N, Einkauf K, Chowdhury FZ, Sun X, Harrington S, Kuo HH, Hua S, Chen HR, Ouyang Z, Reddy K, Dong K, Ndung'u T, Walker BD, Rosenberg ES, Yu XG, Lichterfeld M. Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J Clin Invest 2017. [PMID: 28628034 DOI: 10.1172/jci93289] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
HIV-1 causes a chronic, incurable disease due to its persistence in CD4+ T cells that contain replication-competent provirus, but exhibit little or no active viral gene expression and effectively resist combination antiretroviral therapy (cART). These latently infected T cells represent an extremely small proportion of all circulating CD4+ T cells but possess a remarkable long-term stability and typically persist throughout life, for reasons that are not fully understood. Here we performed massive single-genome, near-full-length next-generation sequencing of HIV-1 DNA derived from unfractionated peripheral blood mononuclear cells, ex vivo-isolated CD4+ T cells, and subsets of functionally polarized memory CD4+ T cells. This approach identified multiple sets of independent, near-full-length proviral sequences from cART-treated individuals that were completely identical, consistent with clonal expansion of CD4+ T cells harboring intact HIV-1. Intact, near-full-genome HIV-1 DNA sequences that were derived from such clonally expanded CD4+ T cells constituted 62% of all analyzed genome-intact sequences in memory CD4 T cells, were preferentially observed in Th1-polarized cells, were longitudinally detected over a duration of up to 5 years, and were fully replication- and infection-competent. Together, these data suggest that clonal proliferation of Th1-polarized CD4+ T cells encoding for intact HIV-1 represents a driving force for stabilizing the pool of latently infected CD4+ T cells.
Collapse
Affiliation(s)
- Guinevere Q Lee
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Nina Orlova-Fink
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Kevin Einkauf
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | | | - Xiaoming Sun
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Sean Harrington
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Hsiao-Hsuan Kuo
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Stephane Hua
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Hsiao-Rong Chen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Zhengyu Ouyang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Kavidha Reddy
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, and
| | - Krista Dong
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, and
| | - Thumbi Ndung'u
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, and.,KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.,Max Planck Institute for Infection Biology, Berlin, Germany
| | - Bruce D Walker
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Eric S Rosenberg
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
81
|
Hosokawa K, Kajigaya S, Keyvanfar K, Qiao W, Xie Y, Townsley DM, Feng X, Young NS. T Cell Transcriptomes from Paroxysmal Nocturnal Hemoglobinuria Patients Reveal Novel Signaling Pathways. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28630090 DOI: 10.4049/jimmunol.1601299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired disorder originating from hematopoietic stem cells and is a life-threating disease characterized by intravascular hemolysis, bone marrow (BM) failure, and venous thrombosis. The etiology of PNH is a somatic mutation in the phosphatidylinositol glycan class A gene (PIG-A) on the X chromosome, which blocks synthesis of the glycolipid moiety and causes deficiency in GPI-anchored proteins. PNH is closely related to aplastic anemia, in which T cells mediate destruction of BM. To identify aberrant molecular mechanisms involved in immune targeting of hematopoietic stem cells in BM, we applied RNA-seq to examine the transcriptome of T cell subsets (CD4+ naive, CD4+ memory, CD8+ naive, and CD8+ memory) from PNH patients and healthy control subjects. Differentially expressed gene analysis in four different T cell subsets from PNH and healthy control subjects showed distinct transcriptional profiles, depending on the T cell subsets. By pathway analysis, we identified novel signaling pathways in T cell subsets from PNH, including increased gene expression involved in TNFR, IGF1, NOTCH, AP-1, and ATF2 pathways. Dysregulation of several candidate genes (JUN, TNFAIP3, TOB1, GIMAP4, GIMAP6, TRMT112, NR4A2, CD69, and TNFSF8) was validated by quantitative real-time RT-PCR and flow cytometry. We have demonstrated molecular signatures associated with positive and negative regulators in T cells, suggesting novel pathophysiologic mechanisms in PNH. These pathways may be targets for new strategies to modulate T cell immune responses in BM failure.
Collapse
Affiliation(s)
- Kohei Hosokawa
- Cell Biology Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Sachiko Kajigaya
- Cell Biology Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Keyvan Keyvanfar
- Cell Biology Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Wangmin Qiao
- Beijing Genomics Institute-Shenzhen, Shenzhen 518083, China
| | - Yanling Xie
- Beijing Genomics Institute-Shenzhen, Shenzhen 518083, China
| | - Danielle M Townsley
- Cell Biology Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Xingmin Feng
- Cell Biology Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Neal S Young
- Cell Biology Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
82
|
Finley J. Elimination of cancer stem cells and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking inhibition of tumorigenesis and the potential eradication of HIV-1. Med Hypotheses 2017; 104:133-146. [PMID: 28673572 DOI: 10.1016/j.mehy.2017.05.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 02/28/2017] [Accepted: 05/26/2017] [Indexed: 12/25/2022]
Abstract
Although promising treatments are currently in development to slow disease progression and increase patient survival, cancer remains the second leading cause of death in the United States. Cancer treatment modalities commonly include chemoradiation and therapies that target components of aberrantly activated signaling pathways. However, treatment resistance is a common occurrence and recent evidence indicates that the existence of cancer stem cells (CSCs) may underlie the limited efficacy and inability of current treatments to effectuate a cure. CSCs, which are largely resistant to chemoradiation therapy, are a subpopulation of cancer cells that exhibit characteristics similar to embryonic stem cells (ESCs), including self-renewal, multi-lineage differentiation, and the ability to initiate tumorigenesis. Interestingly, intracellular mechanisms that sustain quiescence and promote self-renewal in adult stem cells (ASCs) and CSCs likely also function to maintain latency of HIV-1 in CD4+ memory T cells. Although antiretroviral therapy is highly effective in controlling HIV-1 replication, the persistence of latent but replication-competent proviruses necessitates the development of compounds that are capable of selectively reactivating the latent virus, a method known as the "shock and kill" approach. Homeostatic proliferation in central CD4+ memory T (TCM) cells, a memory T cell subset that exhibits limited self-renewal and differentiation and is a primary reservoir for latent HIV-1, has been shown to reinforce and stabilize the latent reservoir in the absence of T cell activation and differentiation. HIV-1 has also been found to establish durable and long-lasting latency in a recently discovered subset of CD4+ T cells known as T memory stem (TSCM) cells. TSCM cells, compared to TCM cells, exhibit stem cell properties that more closely match those of ESCs and ASCs, including self-renewal and differentiation into all memory T cell subsets. It is our hypothesis that activation of AMPK, a master regulator of cellular metabolism that plays a critical role in T cell activation and differentiation of ESCs and ASCs, will lead to both T cell activation-induced latent HIV-1 reactivation, facilitating virus destruction, as well as "activation", differentiation, and/or apoptosis of CSCs, thus inhibiting tumorigenesis. We also propose the novel observation that compounds that have been shown to both facilitate latent HIV-1 reactivation and promote CSC differentiation/apoptosis (e.g. bryostatin-1, JQ1, metformin, butyrate, etc.) likely do so through a common mechanism of AMPK activation.
Collapse
Affiliation(s)
- Jahahreeh Finley
- Finley BioSciences, 9900 Richmond Avenue, #823, Houston, TX 77042-4539, United States.
| |
Collapse
|
83
|
Busby E, Whale AS, Ferns RB, Grant PR, Morley G, Campbell J, Foy CA, Nastouli E, Huggett JF, Garson JA. Instability of 8E5 calibration standard revealed by digital PCR risks inaccurate quantification of HIV DNA in clinical samples by qPCR. Sci Rep 2017; 7:1209. [PMID: 28446770 PMCID: PMC5430807 DOI: 10.1038/s41598-017-01221-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/22/2017] [Indexed: 11/09/2022] Open
Abstract
ABTRACT Establishing a cure for HIV is hindered by the persistence of latently infected cells which constitute the viral reservoir. Real-time qPCR, used for quantification of this reservoir by measuring HIV DNA, requires external calibration; a common choice of calibrator is the 8E5 cell line, which is assumed to be stable and to contain one HIV provirus per cell. In contrast, digital PCR requires no external calibration and potentially provides 'absolute' quantification. We compared the performance of qPCR and dPCR in quantifying HIV DNA in 18 patient samples. HIV DNA was detected in 18 by qPCR and in 15 by dPCR, the difference being due to the smaller sample volume analysed by dPCR. There was good quantitative correlation (R2 = 0.86) between the techniques but on average dPCR values were only 60% of qPCR values. Surprisingly, investigation revealed that this discrepancy was due to loss of HIV DNA from the 8E5 cell calibrant. 8E5 extracts from two other sources were also shown to have significantly less than one HIV DNA copy per cell and progressive loss of HIV from 8E5 cells during culture was demonstrated. We therefore suggest that the copy number of HIV in 8E5 extracts be established by dPCR prior to use as calibrator.
Collapse
Affiliation(s)
- Eloise Busby
- Molecular and Cell Biology Team, LGC, Teddington, UK
| | | | - R Bridget Ferns
- Department of Infection, Division of Infection and Immunity, University College London, London, UK
| | - Paul R Grant
- Department of Clinical Virology, University College London Hospital NHS Foundation Trust, and the UCL/UCLH NIHR Biomedical Research Centre, London, UK
| | - Gary Morley
- Molecular and Cell Biology Team, LGC, Teddington, UK
| | | | - Carole A Foy
- Molecular and Cell Biology Team, LGC, Teddington, UK
| | - Eleni Nastouli
- Department of Clinical Virology, University College London Hospital NHS Foundation Trust, and the UCL/UCLH NIHR Biomedical Research Centre, London, UK.,Department of Population Policy and Practice, UCL GOS Institute of Child Health, London, UK
| | - Jim F Huggett
- Molecular and Cell Biology Team, LGC, Teddington, UK. .,School of Biosciences & Medicine, Faculty of Health & Medical Science, University of Surrey, Guildford, GU2 7XH, UK.
| | - Jeremy A Garson
- Department of Infection, Division of Infection and Immunity, University College London, London, UK. .,National Transfusion Microbiology Laboratories, NHS Blood and Transplant, Colindale, London, UK.
| |
Collapse
|
84
|
Abstract
OBJECTIVE To review the recent literatures related to the factors associated with the size of the HIV reservoir and their clinical significance. DATA SOURCES Literatures related to the size of HIV DNA was collected from PubMed published from 1999 to June 2016. STUDY SELECTION All relevant articles on the HIV DNA and reservoir were collected and reviewed, with no limitation of study design. RESULTS The composition and development of the HIV-1 DNA reservoir in either treated or untreated patients is determined by integrated mechanism comprising viral characteristics, immune system, and treatment strategies. The HIV DNA reservoir is a combination of latency and activity. The residual viremia from the stochastic activation of the reservoir acts as the fuse, continuing to stimulate the immune system to maintain the activated microenvironment for the rebound of competent virus once treatment with antiretroviral therapy is discontinued. CONCLUSION The size of the HIV-1 DNA pool and its composition has great significance in clinical treatment and disease progression.
Collapse
Affiliation(s)
- Ni-Dan Wang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Tai-Sheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
85
|
Gosselin A, Wiche Salinas TR, Planas D, Wacleche VS, Zhang Y, Fromentin R, Chomont N, Cohen ÉA, Shacklett B, Mehraj V, Ghali MP, Routy JP, Ancuta P. HIV persists in CCR6+CD4+ T cells from colon and blood during antiretroviral therapy. AIDS 2017; 31:35-48. [PMID: 27835617 PMCID: PMC5131694 DOI: 10.1097/qad.0000000000001309] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/07/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The objective of this article is to investigate the contribution of colon and blood CD4 T-cell subsets expressing the chemokine receptor CCR6 to HIV persistence during antiretroviral therapy. DESIGN Matched sigmoid biopsies and blood samples (n = 13) as well as leukapheresis (n = 20) were collected from chronically HIV-infected individuals receiving antiretroviral therapy. Subsets of CD4 T cells with distinct differentiation/polarization profiles were identified using surface markers as follows: memory (TM, CD45RA), central memory (TCM; CD45RACCR7), effector (TEM/TM; CD45RACCR7), Th17 (CCR6CCR4), Th1Th17 (CCR6CXCR3), Th1 (CCR6CXCR3), and Th2 (CCR6CCR4). METHODS We used polychromatic flow cytometry for cell sorting, nested real-time PCR for HIV DNA quantification, ELISA and flow cytometry for HIV p24 quantification. HIV reactivation was induced by TCR triggering in the presence/absence of all-trans retinoic acid. RESULTS Compared with blood, the frequency of CCR6 TM was higher in the colon. In both colon and blood compartments, CCR6 TM were significantly enriched in HIV DNA when compared with their CCR6 counterparts (n = 13). In blood, integrated HIV DNA levels were significantly enriched in CCR6 versus CCR6 TCM of four of five individuals and CCR6 versus CCR6 TEM of three of five individuals. Among blood TCM, Th17 and Th1Th17 contributed the most to the pool of cells harboring integrated HIV DNA despite their reduced frequency compared with Th2, which were infected the least. HIV reactivation was induced by TCR triggering and/or retinoic acid exposure at higher levels in CCR6 versus CCR6 TM, TCM, and TEM. CONCLUSION CCR6 is a marker for colon and blood CD4 T cells enriched for replication-competent HIV DNA. Novel eradication strategies should target HIV persistence in CCR6CD4 T cells from various anatomic sites.
Collapse
Affiliation(s)
| | - Tomas Raul Wiche Salinas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Delphine Planas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Vanessa S. Wacleche
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Yuwei Zhang
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | | | - Nicolas Chomont
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Éric A. Cohen
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
- Institut de Recherche Clinique de Montréal, Montréal, Québec, Canada
| | | | - Vikram Mehraj
- Chronic Viral Illness Service and Research Institute
| | | | - Jean-Pierre Routy
- Chronic Viral Illness Service and Research Institute
- Division of Hematology, McGill University Health Centre, Montreal, Québec, Canada
| | - Petronela Ancuta
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| |
Collapse
|
86
|
Ahmed R, Roger L, Costa Del Amo P, Miners KL, Jones RE, Boelen L, Fali T, Elemans M, Zhang Y, Appay V, Baird DM, Asquith B, Price DA, Macallan DC, Ladell K. Human Stem Cell-like Memory T Cells Are Maintained in a State of Dynamic Flux. Cell Rep 2016; 17:2811-2818. [PMID: 27974195 PMCID: PMC5186732 DOI: 10.1016/j.celrep.2016.11.037] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/23/2016] [Accepted: 11/10/2016] [Indexed: 11/29/2022] Open
Abstract
Adaptive immunity requires the generation of memory T cells from naive precursors selected in the thymus. The key intermediaries in this process are stem cell-like memory T (TSCM) cells, multipotent progenitors that can both self-renew and replenish more differentiated subsets of memory T cells. In theory, antigen specificity within the TSCM pool may be imprinted statically as a function of largely dormant cells and/or retained dynamically by more transitory subpopulations. To explore the origins of immunological memory, we measured the turnover of TSCM cells in vivo using stable isotope labeling with heavy water. The data indicate that TSCM cells in both young and elderly subjects are maintained by ongoing proliferation. In line with this finding, TSCM cells displayed limited telomere length erosion coupled with high expression levels of active telomerase and Ki67. Collectively, these observations show that TSCM cells exist in a state of perpetual flux throughout the human lifespan.
Collapse
Affiliation(s)
- Raya Ahmed
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Laureline Roger
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Pedro Costa Del Amo
- Department of Medicine, St. Mary's Hospital, Imperial College London, London W2 1PG, UK
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Rhiannon E Jones
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Lies Boelen
- Department of Medicine, St. Mary's Hospital, Imperial College London, London W2 1PG, UK
| | - Tinhinane Fali
- Sorbonne Universités, UPMC Université Paris 06, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France; INSERM U1135, CIMI-Paris, 75013 Paris, France
| | - Marjet Elemans
- Department of Medicine, St. Mary's Hospital, Imperial College London, London W2 1PG, UK
| | - Yan Zhang
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Victor Appay
- Sorbonne Universités, UPMC Université Paris 06, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France; INSERM U1135, CIMI-Paris, 75013 Paris, France
| | - Duncan M Baird
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Becca Asquith
- Department of Medicine, St. Mary's Hospital, Imperial College London, London W2 1PG, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Derek C Macallan
- Institute for Infection and Immunity, St. George's, University of London, London SW17 0RE, UK; St. George's University Hospitals National Health Service Foundation Trust, Blackshaw Road, London SW17 0QT, UK.
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
87
|
Tsunetsugu-Yokota Y, Kobayahi-Ishihara M, Wada Y, Terahara K, Takeyama H, Kawana-Tachikawa A, Tokunaga K, Yamagishi M, Martinez JP, Meyerhans A. Homeostatically Maintained Resting Naive CD4 + T Cells Resist Latent HIV Reactivation. Front Microbiol 2016; 7:1944. [PMID: 27990142 PMCID: PMC5130990 DOI: 10.3389/fmicb.2016.01944] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 11/18/2016] [Indexed: 02/03/2023] Open
Abstract
Homeostatic proliferation (HSP) is a major mechanism by which long-lived naïve and memory CD4+ T cells are maintained in vivo and suggested to contribute to the persistence of the latent HIV-1 reservoir. However, while many in vitro latency models rely on CD4+ T cells that were initially differentiated via T-cell receptor (TCR) stimulation into memory/effector cells, latent infection of naïve resting CD4+ T cells maintained under HSP conditions has not been fully addressed. Here, we describe an in vitro HSP culture system utilizing the cytokines IL-7 and IL-15 that allows studying latency in naïve resting CD4+ T cells. CD4+ T cells isolated from several healthy donors were infected with HIV pseudotypes expressing GFP and cultured under HSP conditions or TCR conditions as control. Cell proliferation, phenotype, and GFP expression were analyzed by flow cytometry. RNA expression was quantified by qRT-PCR. Under HSP culture conditions, latently HIV-1 infected naïve cells are in part maintained in the non-dividing (= resting) state. Although a few HIV-1 provirus+ cells were present in these resting GFP negative cells, the estimated level of GFP transcripts per infected cell seems to indicate a block at the post-transcriptional level. Interestingly, neither TCR nor the prototypic HDAC inhibitor SAHA were able to reactivate HIV-1 provirus from these cells. This lack of reactivation was not due to methylation of the HIV LTR. These results point to a mechanism of HIV control in HSP-cultured resting naïve CD4+ T cells that may be distinct from that in TCR-stimulated memory/effector T cells.
Collapse
Affiliation(s)
- Yasuko Tsunetsugu-Yokota
- Department of Medical Technology, School of Human Sciences, Tokyo University of TechnologyTokyo, Japan; Department of Immunology, National Institute of Infectious DiseasesTokyo, Japan
| | | | - Yamato Wada
- Department of Immunology, National Institute of Infectious DiseasesTokyo, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda UniversityTokyo, Japan
| | - Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases Tokyo, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University Tokyo, Japan
| | - Ai Kawana-Tachikawa
- AIDS Research Center, National Institute of Infectious Diseases Tokyo, Japan
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases Tokyo, Japan
| | - Makoto Yamagishi
- Graduate School of Frontier Sciences, University of Tokyo Tokyo, Japan
| | - Javier P Martinez
- Infection Biology Group, Department of Experimental and Health Sciences, University Pompeu Fabra Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Group, Department of Experimental and Health Sciences, University Pompeu FabraBarcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona, Spain
| |
Collapse
|
88
|
Duvigneau S, Sharma-Chawla N, Boianelli A, Stegemann-Koniszewski S, Nguyen VK, Bruder D, Hernandez-Vargas EA. Hierarchical effects of pro-inflammatory cytokines on the post-influenza susceptibility to pneumococcal coinfection. Sci Rep 2016; 6:37045. [PMID: 27872472 PMCID: PMC5181841 DOI: 10.1038/srep37045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/20/2016] [Indexed: 02/07/2023] Open
Abstract
In the course of influenza A virus (IAV) infections, a secondary bacterial infection frequently leads to serious respiratory conditions provoking high hospitalization and death tolls. Although abundant pro-inflammatory responses have been reported as key contributing factors for these severe dual infections, the relative contributions of cytokines remain largely unclear. In the current study, mathematical modelling based on murine experimental data dissects IFN-γ as a cytokine candidate responsible for impaired bacterial clearance, thereby promoting bacterial growth and systemic dissemination during acute IAV infection. We also found a time-dependent detrimental role of IL-6 in curtailing bacterial outgrowth which was not as distinct as for IFN-γ. Our numerical simulations suggested a detrimental effect of IFN-γ alone and in synergism with IL-6 but no conclusive pathogenic effect of IL-6 and TNF-α alone. This work provides a rationale to understand the potential impact of how to manipulate temporal immune components, facilitating the formulation of hypotheses about potential therapeutic strategies to treat coinfections.
Collapse
Affiliation(s)
- Stefanie Duvigneau
- Infection Immunology Group, Institute of Medical Microbiology, Disease Prevention and Control, Otto-von-Guericke University Magdeburg, Germany.,Systems Medicine of Infectious Disease Group, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Chair for Automation/Modeling, Institute for Automation Engineering, Otto-von-Guericke University Magdeburg, Germany
| | - Niharika Sharma-Chawla
- Infection Immunology Group, Institute of Medical Microbiology, Disease Prevention and Control, Otto-von-Guericke University Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Alessandro Boianelli
- Systems Medicine of Infectious Disease Group, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sabine Stegemann-Koniszewski
- Infection Immunology Group, Institute of Medical Microbiology, Disease Prevention and Control, Otto-von-Guericke University Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Van Kinh Nguyen
- Systems Medicine of Infectious Disease Group, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Disease Prevention and Control, Otto-von-Guericke University Magdeburg, Germany.,Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Esteban A Hernandez-Vargas
- Systems Medicine of Infectious Disease Group, Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
89
|
Multi-dose Romidepsin Reactivates Replication Competent SIV in Post-antiretroviral Rhesus Macaque Controllers. PLoS Pathog 2016; 12:e1005879. [PMID: 27632364 PMCID: PMC5025140 DOI: 10.1371/journal.ppat.1005879] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/18/2016] [Indexed: 11/19/2022] Open
Abstract
Viruses that persist despite seemingly effective antiretroviral treatment (ART) and can reinitiate infection if treatment is stopped preclude definitive treatment of HIV-1 infected individuals, requiring lifelong ART. Among strategies proposed for targeting these viral reservoirs, the premise of the “shock and kill” strategy is to induce expression of latent proviruses [for example with histone deacetylase inhibitors (HDACis)] resulting in elimination of the affected cells through viral cytolysis or immune clearance mechanisms. Yet, ex vivo studies reported that HDACis have variable efficacy for reactivating latent proviruses, and hinder immune functions. We developed a nonhuman primate model of post-treatment control of SIV through early and prolonged administration of ART and performed in vivo reactivation experiments in controller RMs, evaluating the ability of the HDACi romidepsin (RMD) to reactivate SIV and the impact of RMD treatment on SIV-specific T cell responses. Ten RMs were IV-infected with a SIVsmmFTq transmitted-founder infectious molecular clone. Four RMs received conventional ART for >9 months, starting from 65 days post-infection. SIVsmmFTq plasma viremia was robustly controlled to <10 SIV RNA copies/mL with ART, without viral blips. At ART cessation, initial rebound viremia to ~106 copies/mL was followed by a decline to < 10 copies/mL, suggesting effective immune control. Three post-treatment controller RMs received three doses of RMD every 35–50 days, followed by in vivo experimental depletion of CD8+ cells using monoclonal antibody M-T807R1. RMD was well-tolerated and resulted in a rapid and massive surge in T cell activation, as well as significant virus rebounds (~104 copies/ml) peaking at 5–12 days post-treatment. CD8+ cell depletion resulted in a more robust viral rebound (107 copies/ml) that was controlled upon CD8+ T cell recovery. Our results show that RMD can reactivate SIV in vivo in the setting of post-ART viral control. Comparison of the patterns of virus rebound after RMD administration and CD8+ cell depletion suggested that RMD impact on T cells is only transient and does not irreversibly alter the ability of SIV-specific T cells to control the reactivated virus. Antiretroviral therapy (ART) does not eradicate HIV-1 in infected individuals due to virus persistence in latently infected reservoir cells, despite apparently effective ART. The persistent virus and can rekindle infection when ART is interrupted. The goal of the “shock and kill” viral clearance strategy is to induce expression of latent proviruses and eliminate the infected cells through viral cytolysis or immune clearance mechanisms. Latency reversing agents (LRAs) tested to date have been reported to have variable effects, both on virus reactivation and on immune functions. We performed in vivo reactivation experiments in SIV-infected RMs that controlled viral replication after a period of ART to evaluate the ability of the histone deacetylase inhibitor romidepsin (RMD) to reactivate SIV and its impact on SIV-specific immune responses. Our results suggest that RMD treatment can increase virus expression in this setting, and that it does not markedly or durably impair the ability of SIV-specific T cells to control viral replication.
Collapse
|
90
|
|
91
|
Fromentin R, Bakeman W, Lawani MB, Khoury G, Hartogensis W, DaFonseca S, Killian M, Epling L, Hoh R, Sinclair E, Hecht FM, Bacchetti P, Deeks SG, Lewin SR, Sékaly RP, Chomont N. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART. PLoS Pathog 2016; 12:e1005761. [PMID: 27415008 PMCID: PMC4944956 DOI: 10.1371/journal.ppat.1005761] [Citation(s) in RCA: 338] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022] Open
Abstract
HIV persists in a small pool of latently infected cells despite antiretroviral therapy (ART). Identifying cellular markers expressed at the surface of these cells may lead to novel therapeutic strategies to reduce the size of the HIV reservoir. We hypothesized that CD4+ T cells expressing immune checkpoint molecules would be enriched in HIV-infected cells in individuals receiving suppressive ART. Expression levels of 7 immune checkpoint molecules (PD-1, CTLA-4, LAG-3, TIGIT, TIM-3, CD160 and 2B4) as well as 4 markers of HIV persistence (integrated and total HIV DNA, 2-LTR circles and cell-associated unspliced HIV RNA) were measured in PBMCs from 48 virally suppressed individuals. Using negative binomial regression models, we identified PD-1, TIGIT and LAG-3 as immune checkpoint molecules positively associated with the frequency of CD4+ T cells harboring integrated HIV DNA. The frequency of CD4+ T cells co-expressing PD-1, TIGIT and LAG-3 independently predicted the frequency of cells harboring integrated HIV DNA. Quantification of HIV genomes in highly purified cell subsets from blood further revealed that expressions of PD-1, TIGIT and LAG-3 were associated with HIV-infected cells in distinct memory CD4+ T cell subsets. CD4+ T cells co-expressing the three markers were highly enriched for integrated viral genomes (median of 8.2 fold compared to total CD4+ T cells). Importantly, most cells carrying inducible HIV genomes expressed at least one of these markers (median contribution of cells expressing LAG-3, PD-1 or TIGIT to the inducible reservoir = 76%). Our data provide evidence that CD4+ T cells expressing PD-1, TIGIT and LAG-3 alone or in combination are enriched for persistent HIV during ART and suggest that immune checkpoint blockers directed against these receptors may represent valuable tools to target latently infected cells in virally suppressed individuals.
Collapse
Affiliation(s)
- Rémi Fromentin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Wendy Bakeman
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
| | - Mariam B. Lawani
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
| | - Gabriela Khoury
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Wendy Hartogensis
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Sandrina DaFonseca
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
| | - Marisela Killian
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Lorrie Epling
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Elizabeth Sinclair
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Frederick M. Hecht
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Sharon R. Lewin
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | | | - Nicolas Chomont
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
92
|
Initiation of Antiretroviral Therapy Restores CD4+ T Memory Stem Cell Homeostasis in Simian Immunodeficiency Virus-Infected Macaques. J Virol 2016; 90:6699-6708. [PMID: 27170752 DOI: 10.1128/jvi.00492-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/06/2016] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Treatment of human immunodeficiency virus (HIV) infection with antiretroviral therapy (ART) has significantly improved prognosis. Unfortunately, interruption of ART almost invariably results in viral rebound, attributed to a pool of long-lived, latently infected cells. Based on their longevity and proliferative potential, CD4(+) T memory stem cells (TSCM) have been proposed as an important site of HIV persistence. In a previous study, we found that in simian immunodeficiency virus (SIV)-infected rhesus macaques (RM), CD4(+) TSCM are preserved in number but show (i) a decrease in the frequency of CCR5(+) cells, (ii) an expansion of the fraction of proliferating Ki-67(+) cells, and (iii) high levels of SIV DNA. To understand the impact of ART on both CD4(+) TSCM homeostasis and virus persistence, we conducted a longitudinal analysis of these cells in the blood and lymph nodes of 25 SIV-infected RM. We found that ART induced a significant restoration of CD4(+) CCR5(+) TSCM both in blood and in lymph nodes and a reduction in the fraction of proliferating CD4(+) Ki-67(+) TSCM in blood (but not lymph nodes). Importantly, we found that the level of SIV DNA in CD4(+) transitional memory (TTM) and effector memory (TEM) T cells declined ∼100-fold after ART in both blood and lymph nodes, while the level of SIV DNA in CD4(+) TSCM and central memory T cells (TCM-) did not significantly change. These data suggest that ART is effective at partially restoring CD4(+) TSCM homeostasis, and the observed stable level of virus in TSCM supports the hypothesis that these cells are a critical contributor to SIV persistence. IMPORTANCE Understanding the roles of various CD4(+) T cell memory subsets in immune homeostasis and HIV/SIV persistence during antiretroviral therapy (ART) is critical to effectively treat and cure HIV infection. T memory stem cells (TSCM) are a unique memory T cell subset with enhanced self-renewal capacity and the ability to differentiate into other memory T cell subsets, such as central and transitional memory T cells (TCM and TTM, respectively). CD4(+) TSCM are disrupted but not depleted during pathogenic SIV infection. We find that ART is partially effective at restoring CD4(+) TSCM homeostasis and that SIV DNA harbored within this subset contracts more slowly than virus harbored in shorter-lived subsets, such as TTM and effector memory (TEM). Because of their ability to persist long-term in an individual, understanding the dynamics of virally infected CD4(+) TSCM during suppressive ART is important for future therapeutic interventions aimed at modulating immune activation and purging the HIV reservoir.
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW The introduction of effective antiretroviral therapy (ART) has transformed HIV infection from a deadly to a chronic infection. Despite its successes in reducing mortality, ART fails to cure HIV allowing HIV to persist in vivo. HIV persistence under ART is thought to be mediated by a combination of latent infection of long-lived cells, homeostatic proliferation of latently infected cells, anatomic sanctuaries, and low-level virus replication. To understand the contribution of specific cell types and anatomic sites to virus persistence in vivo animal models are necessary. RECENT FINDINGS The advancements in ART and our understanding of animal models have facilitated the development of models of HIV persistence in nonhuman primates and mice. Simian immunodeficiency virus (SIV) or simian/HIV infection (SHIV) of rhesus and pigtail macaques followed by effective ART represents the most faithful animal model of HIV persistence. HIV infection of humanized mice also provides a useful model for answering specific questions regarding virus persistence in a uniquely mutable system. SUMMARY In this review, we describe the most recent findings using animal models of HIV persistence. We will first describe the important aspects of HIV infection that SIV/SHIV infection of nonhuman primates are able to recapitulate, then we will discuss some recent studies that have used these models to understand viral persistence.
Collapse
|
94
|
Cummins NW, Sainski AM, Dai H, Natesampillai S, Pang YP, Bren GD, de Araujo Correia MCM, Sampath R, Rizza SA, O'Brien D, Yao JD, Kaufmann SH, Badley AD. Prime, Shock, and Kill: Priming CD4 T Cells from HIV Patients with a BCL-2 Antagonist before HIV Reactivation Reduces HIV Reservoir Size. J Virol 2016; 90:4032-4048. [PMID: 26842479 PMCID: PMC4810548 DOI: 10.1128/jvi.03179-15] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/28/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Understanding how some HIV-infected cells resist the cytotoxicity of HIV replication is crucial to enabling HIV cure efforts. HIV killing of CD4 T cells that replicate HIV can involve HIV protease-mediated cleavage of procaspase 8 to generate a fragment (Casp8p41) that directly binds and activates the mitochondrial proapoptotic protein BAK. Here, we demonstrate that Casp8p41 also binds with nanomolar affinity to the antiapoptotic protein Bcl-2, which sequesters Casp8p41 and prevents apoptosis. Further, we show that central memory CD4 T cells (TCM) from HIV-infected individuals have heightened expression of BCL-2 relative to procaspase 8, possibly explaining the persistence of HIV-infected TCMdespite generation of Casp8p41. Consistent with this hypothesis, the selective BCL-2 antagonist venetoclax induced minimal killing of uninfected CD4 T cells but markedly increased the death of CD4 T cells and diminished cell-associated HIV DNA when CD4 T cells from antiretroviral therapy (ART)-suppressed HIV patients were induced with αCD3/αCD28 to reactivate HIVex vivo Thus, priming CD4 T cells from ART suppressed HIV patients with a BCL-2 antagonist, followed by HIV reactivation, achieves reductions in cell-associated HIV DNA, whereas HIV reactivation alone does not. IMPORTANCE HIV infection is incurable due to a long-lived reservoir of HIV(+)memory CD4 T cells, and no clinically relevant interventions have been identified that reduce the number of these HIV DNA-containing cells. Since postintegration HIV replication can result in HIV protease generation of Casp8p41, which activates BAK, causing infected CD4 T cell death, we sought to determine whether this occurs in memory CD4 T cells. Here, we demonstrate that memory CD4 T cells can generate Casp8p41 and yet are intrinsically resistant to death induced by diverse stimuli, including Casp8p41. Furthermore, BCL-2 expression is relatively increased in these cells and directly binds and inhibits Casp8p41's proapoptotic effects. Antagonizing BCL-2 with venetoclax derepresses this antagonism, resulting in death, preferentially in HIV DNA containing cells, since only these cells generate Casp8p41. Thus, BCL-2 antagonism is a clinically relevant intervention with the potential to reduce HIV reservoir size in patients.
Collapse
Affiliation(s)
- Nathan W Cummins
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Amy M Sainski
- Department of Pharmacology, Mayo Clinic, Rochester, Minnesota, USA
| | - Haiming Dai
- Department of Pharmacology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Yuan-Ping Pang
- Department of Pharmacology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gary D Bren
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Rahul Sampath
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Stacey A Rizza
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel O'Brien
- Department of Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Joseph D Yao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott H Kaufmann
- Department of Pharmacology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
95
|
Abstract
Recent anecdotal reports of HIV-infected children who received early antiretroviral therapy (ART) and showed sustained control of viral replication even after ART discontinuation have raised the question of whether there is greater intrinsic potential for HIV remission, or even eradication ('cure'), in paediatric infection than in adult infection. This Review describes the influence of early initiation of ART, of immune ontogeny and of maternal factors on the potential for HIV cure in children and discusses the unique immunotherapeutic opportunities and obstacles that paediatric infection may present.
Collapse
Affiliation(s)
- Philip J Goulder
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, UK
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne 3004, Australia
| | - Ellen M Leitman
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, UK
| |
Collapse
|
96
|
Policicchio BB, Pandrea I, Apetrei C. Animal Models for HIV Cure Research. Front Immunol 2016; 7:12. [PMID: 26858716 PMCID: PMC4729870 DOI: 10.3389/fimmu.2016.00012] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/12/2016] [Indexed: 12/17/2022] Open
Abstract
The HIV-1/AIDS pandemic continues to spread unabated worldwide, and no vaccine exists within our grasp. Effective antiretroviral therapy (ART) has been developed, but ART cannot clear the virus from the infected patient. A cure for HIV-1 is badly needed to stop both the spread of the virus in human populations and disease progression in infected individuals. A safe and effective cure strategy for human immunodeficiency virus (HIV) infection will require multiple tools, and appropriate animal models are tools that are central to cure research. An ideal animal model should recapitulate the essential aspects of HIV pathogenesis and associated immune responses, while permitting invasive studies, thus allowing a thorough evaluation of strategies aimed at reducing the size of the reservoir (functional cure) or eliminating the reservoir altogether (sterilizing cure). Since there is no perfect animal model for cure research, multiple models have been tailored and tested to address specific quintessential questions of virus persistence and eradication. The development of new non-human primate and mouse models, along with a certain interest in the feline model, has the potential to fuel cure research. In this review, we highlight the major animal models currently utilized for cure research and the contributions of each model to this goal.
Collapse
Affiliation(s)
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh , Pittsburgh, PA , USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
97
|
Chahroudi A, Silvestri G, Lichterfeld M. T memory stem cells and HIV: a long-term relationship. Curr HIV/AIDS Rep 2016; 12:33-40. [PMID: 25578055 DOI: 10.1007/s11904-014-0246-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In analogy to many tissues in which mature, terminally differentiated cells are continuously replenished by the progeny of less differentiated, long-lasting stem cells, it has been suspected that memory T lymphocytes might contain small numbers of stem cell-like cells. However, only recently have such cells been physically identified and isolated from humans, mice, and nonhuman primates. These cells, termed "T memory stem cells" (TSCM), represent approximately 2-4 % of all circulating T lymphocytes, seem to be extremely durable, and can rapidly differentiate into more mature central memory, effector memory, and effector T cells, while maintaining their own pool size through homeostatic self-renewal. Although it is becoming increasingly evident that that these cells have critical roles for T cell homeostasis and maintaining life-long cellular immunity against microbial pathogens during physiological conditions, they also seem intrinsically involved in many key aspects of HIV/SIV disease pathogenesis. Current data suggest that CD4+ TSCM cells represent a core element of the HIV-1 reservoir in patients treated with suppressive antiretroviral therapy (ART) and that relative resistance of CD4+ TSCM cells to SIV represents a distinguishing feature of non-pathogenic SIV infection in natural hosts. This article summarizes recent studies investigating the role of TSCM in HIV/SIV infection.
Collapse
Affiliation(s)
- Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA,
| | | | | |
Collapse
|
98
|
Hosokawa K, Muranski P, Feng X, Townsley DM, Liu B, Knickelbein J, Keyvanfar K, Dumitriu B, Ito S, Kajigaya S, Taylor JG, Kaplan MJ, Nussenblatt RB, Barrett AJ, O'Shea J, Young NS. Memory Stem T Cells in Autoimmune Disease: High Frequency of Circulating CD8+ Memory Stem Cells in Acquired Aplastic Anemia. THE JOURNAL OF IMMUNOLOGY 2016; 196:1568-78. [PMID: 26764034 DOI: 10.4049/jimmunol.1501739] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/08/2015] [Indexed: 11/19/2022]
Abstract
Memory stem T cells (TSCMs) constitute a long-lived, self-renewing lymphocyte population essential for the maintenance of functional immunity. Hallmarks of autoimmune disease pathogenesis are abnormal CD4(+) and CD8(+) T cell activation. We investigated the TSCM subset in 55, 34, 43, and 5 patients with acquired aplastic anemia (AA), autoimmune uveitis, systemic lupus erythematosus, and sickle cell disease, respectively, as well as in 41 age-matched healthy controls. CD8(+) TSCM frequency was significantly increased in AA compared with healthy controls. An increased CD8(+) TSCM frequency at diagnosis was associated with responsiveness to immunosuppressive therapy, and an elevated CD8(+) TSCM population after immunosuppressive therapy correlated with treatment failure or relapse in AA patients. IFN-γ and IL-2 production was significantly increased in various CD8(+) and CD4(+) T cell subsets in AA patients, including CD8(+) and CD4(+) TSCMs. CD8(+) TSCM frequency was also increased in patients with autoimmune uveitis or sickle cell disease. A positive correlation between CD4(+) and CD8(+) TSCM frequencies was found in AA, autoimmune uveitis, and systemic lupus erythematosus. Evaluation of PD-1, CD160, and CD244 expression revealed that TSCMs were less exhausted compared with other types of memory T cells. Our results suggest that the CD8(+) TSCM subset is a novel biomarker and a potential therapeutic target for AA.
Collapse
Affiliation(s)
- Kohei Hosokawa
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892;
| | - Pawel Muranski
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Danielle M Townsley
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Baoying Liu
- Clinical Immunology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jared Knickelbein
- Clinical Immunology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Keyvan Keyvanfar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Bogdan Dumitriu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Sawa Ito
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Sachiko Kajigaya
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - James G Taylor
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Robert B Nussenblatt
- Clinical Immunology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - A John Barrett
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - John O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
99
|
Timilsina U, Gaur R. Modulation of apoptosis and viral latency - an axis to be well understood for successful cure of human immunodeficiency virus. J Gen Virol 2016; 97:813-824. [PMID: 26764023 DOI: 10.1099/jgv.0.000402] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human immunodeficiency virus (HIV) is the causative agent of the deadly disease AIDS, which is characterized by the progressive decline of CD4(+)T-cells. HIV-1-encoded proteins such as envelope gp120 (glycoprotein gp120), Tat (trans-activator of transcription), Nef (negative regulatory factor), Vpr (viral protein R), Vpu (viral protein unique) and protease are known to be effective in modulating host cell signalling pathways that lead to an alteration in apoptosis of both HIV-infected and uninfected bystander cells. Depending on the stage of the virus life cycle and host cell type, these viral proteins act as mediators of pro- or anti-apoptotic signals. HIV latency in viral reservoirs is a persistent phenomenon that has remained beyond the control of the human immune system. To cure HIV infections completely, it is crucial to reactivate latent HIV from cellular pools and to drive these apoptosis-resistant cells towards death. Several previous studies have reported the role of HIV-encoded proteins in apoptosis modulation, but the molecular basis for apoptosis evasion of some chronically HIV-infected cells and reactivated latently HIV-infected cells still needs to be elucidated. The current review summarizes our present understanding of apoptosis modulation in HIV-infected cells, uninfected bystander cells and latently infected cells, with a focus on highlighting strategies to activate the apoptotic pathway to kill latently infected cells.
Collapse
Affiliation(s)
- Uddhav Timilsina
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi- 110021, India
| | - Ritu Gaur
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi- 110021, India
| |
Collapse
|
100
|
Flynn JK, Gorry PR. T cell therapies-are T memory stem cells the answer? ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:251. [PMID: 26605297 DOI: 10.3978/j.issn.2305-5839.2015.08.13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
T memory stem cells (TSCM) are the earliest developmental stage of memory T cells, displaying stem cell-like properties and exhibiting a gene profile between naive and central memory (CM) T cells. Their long-lifespan, robust proliferative potential and self-renewal capacity has generated much research and clinical interest particularly for therapeutic use. Here, we discuss recent findings published in Science Translational Medicine by Biasco and colleagues [2015 Feb 4;7(273):273ra13], which provided evidence for the persistence of TSCM in humans for up to 12 years after infusion of genetically modified lymphocytes, and we examine the implications for the development of novel immunotherapies using TSCM.
Collapse
Affiliation(s)
- Jacqueline K Flynn
- 1 School of Applied Sciences and Program in Metabolism, Exercise and Disease, Health Initiatives Research Institute, RMIT University, Melbourne, Australia ; 2 Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia ; 3 Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia ; 4 Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Paul R Gorry
- 1 School of Applied Sciences and Program in Metabolism, Exercise and Disease, Health Initiatives Research Institute, RMIT University, Melbourne, Australia ; 2 Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia ; 3 Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia ; 4 Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|