1
|
Rueger S, Gruener E, Wang D, Abdool FS, Ober V, Vallée T, Stirner R, Conca R, Andrä I, Rogers L, Zahn R, Gersbacher E, Eger J, Pauli R, Postel N, Spinner CD, Vehreschild JJ, Stecher M, Nitschko H, Eberle J, Bogner JR, Seybold U, Draenert R, Leslie A, Kløverpris HN, Geldmacher C, Muenchhoff M, Held K, Roider J. Early treatment and PD1 inhibition enhance HIV-specific functionality of follicular CD8+ T cells. JCI Insight 2025; 10:e180309. [PMID: 40197363 PMCID: PMC11981630 DOI: 10.1172/jci.insight.180309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/18/2025] [Indexed: 04/10/2025] Open
Abstract
People living with HIV treated during acute infection are the group for whom achieving functional cure appears most viable. Follicular CD8+ T cells could contribute to HIV reservoir clearance by accessing B cell follicles through CXCR5 expression. This study examines peripheral follicular CD8+ T cells using flow cytometry, transcriptome analyses, and functional assays in people treated during acute (n = 37) and chronic (n = 18) infection, as well as in individuals naturally controlling HIV (n = 20) and living without HIV (n = 10). Our results reveal that early, as opposed to late, treatment initiation preserves antiviral effector functions of follicular CD8+ T cells, which are further enhanced by PD1 inhibition. We also identify a correlation between follicular CD8+ T cells and intact proviral HIV DNA levels in acute, but not chronic, infection. Longitudinal transcriptomic analysis of peripheral effector cells after 48 weeks of suppressive therapy indicated traits of recent antigen exposure, suggesting potential recirculation into lymphoid tissue. These findings underscore the pivotal role of follicular CD8+ T cells in anti-HIV responses and support investigating targeted cure strategies, such as anti-PD1 therapy, especially in individuals initiating treatment during acute infection.
Collapse
Affiliation(s)
- Susanne Rueger
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Germany
| | - Eva Gruener
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Danni Wang
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Faiaz Shaik Abdool
- Africa Health Research Institute (AHRI), and
- Department of Laboratory Medicine and Medical Science, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Veronica Ober
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Theresa Vallée
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Renate Stirner
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Raffaele Conca
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, LMU University Hospital, LMU Munich, Munich, Germany
| | - Immanuel Andrä
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Lisa Rogers
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Zahn
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Joanna Eger
- Zentrum fuer Innere Medizin und Infektiologie, Munich, Germany
| | | | - Nils Postel
- prinzmed, Practice for Infectiology, Munich, Germany
| | - Christoph D. Spinner
- TUM School of Medicine and Health, Department of Clinical Medicine – Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Jörg J. Vehreschild
- Medical Department 2, Hematology/Oncology and Infectious Diseases, University Hospital of Frankfurt, Frankfurt, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Melanie Stecher
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, Cologne, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Germany
| | - Hans Nitschko
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Josef Eberle
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Johannes R. Bogner
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Germany
| | - Ulrich Seybold
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rika Draenert
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Antibiotic Stewardship Team, LMU University Hospital, LMU Munich, Munich, Germany
| | - Al Leslie
- Africa Health Research Institute (AHRI), and
- Department of Infection and Immunity, University College London (UCL), London, United Kingdom
| | - Henrik N. Kløverpris
- Africa Health Research Institute (AHRI), and
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Christof Geldmacher
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
| | - Maximilian Muenchhoff
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, and
| | - Kathrin Held
- German Centre for Infection Research (DZIF), partner site Munich, Germany
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
- Unit Global Health, Helmholtz Zentrum München, German Research Centre for Environmental Health (HMGU), Neuherberg, Germany
| | - Julia Roider
- Department of Infectious Diseases, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Germany
| |
Collapse
|
2
|
Nag M, Fogle JE, Pillay S, Del Prete GQ, De Paris K. Tissue-Specific DNA Methylation Changes in CD8 + T Cells During Chronic Simian Immunodeficiency Virus Infection of Infant Rhesus Macaques. Viruses 2024; 16:1839. [PMID: 39772149 PMCID: PMC11680437 DOI: 10.3390/v16121839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Robust CD8+ T cell responses are critical for the control of HIV infection in both adults and children. Our understanding of the mechanisms driving these responses is based largely on studies of cells circulating in peripheral blood in adults, but the regulation of CD8+ T cell responses in tissue sites is poorly understood, particularly in pediatric infections. DNA methylation is an epigenetic modification that regulates gene transcription. Hypermethylated gene promoters are associated with transcriptional silencing and, conversely, hypomethylated promoters indicate gene activation. In this study, we evaluated DNA methylation signatures of CD8+ T cells isolated from several different anatomic compartments during pediatric AIDS-virus infection by utilizing the SIVmac239/251 infected infant rhesus macaque model. We performed a stepwise methylation analysis starting with total cellular DNA, to immunomodulatory cytokine promoters, to specific CpG sites within the cytokine promoters in CD8+ T cells isolated from peripheral blood, lymph nodes, and intestinal tissue during the chronic phase of infection. Tissue-specific methylation patterns were determined for transcriptionally active promoters of key immunomodulatory cytokines: interferon gamma (IFNγ), interleukin-2 (IL-2), and tumor necrosis factor alpha (TNFα). In this study, we observed tissue-specific differences in CD8+ T cell modulation by DNA methylation in SIV-infected infant macaques, highlighting the importance of evaluating cells from both blood and tissues to obtain a complete picture of CD8+ T cell regulation during pediatric HIV infection.
Collapse
Affiliation(s)
- Mukta Nag
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA (K.D.P.)
| | | | - Santhoshan Pillay
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA (K.D.P.)
| | - Gregory Q. Del Prete
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA (K.D.P.)
| |
Collapse
|
3
|
Mainou E, Berendam SJ, Obregon-Perko V, Uffman EA, Phan CT, Shaw GM, Bar KJ, Kumar MR, Fray EJ, Siliciano JM, Siliciano RF, Silvestri G, Permar SR, Fouda GG, McCarthy J, Chahroudi A, Conway JM, Chan C. Assessing the impact of autologous virus neutralizing antibodies on viral rebound time in postnatally SHIV-infected ART-treated infant rhesus macaques. Epidemics 2024; 48:100780. [PMID: 38964130 PMCID: PMC11518701 DOI: 10.1016/j.epidem.2024.100780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/23/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
While the benefits of early antiretroviral therapy (ART) initiation in perinatally infected infants are well documented, early initiation is not always possible in postnatal pediatric HIV infections. The timing of ART initiation is likely to affect the size of the latent viral reservoir established, as well as the development of adaptive immune responses, such as the generation of neutralizing antibody responses against the virus. How these parameters impact the ability of infants to control viremia and the time to viral rebound after ART interruption is unclear and has never been modeled in infants. To investigate this question we used an infant nonhuman primate Simian/Human Immunodeficiency Virus (SHIV) infection model. Infant Rhesus macaques (RMs) were orally challenged with SHIV.C.CH505 375H dCT and either given ART at 4-7 days post-infection (early ART condition), at 2 weeks post-infection (intermediate ART condition), or at 8 weeks post-infection (late ART condition). These infants were then monitored for up to 60 months post-infection with serial viral load and immune measurements. To gain insight into early after analytic treatment interruption (ATI), we constructed mathematical models to investigate the effect of time of ART initiation in delaying viral rebound when treatment is interrupted, focusing on the relative contributions of latent reservoir size and autologous virus neutralizing antibody responses. We developed a stochastic mathematical model to investigate the joint effect of latent reservoir size, the autologous neutralizing antibody potency, and CD4+ T cell levels on the time to viral rebound for RMs rebounding up to 60 days post-ATI. We find that the latent reservoir size is an important determinant in explaining time to viral rebound in infant macaques by affecting the growth rate of the virus. The presence of neutralizing antibodies can also delay rebound, but we find this effect for high potency antibody responses only. Finally, we discuss the therapeutic implications of our findings.
Collapse
Affiliation(s)
- Ellie Mainou
- Department of Biology, Pennsylvania State University, University Park, PA, USA.
| | | | | | - Emilie A Uffman
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Caroline T Phan
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - George M Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharine J Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mithra R Kumar
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily J Fray
- Department of Biochemistry and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janet M Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | | | - Janice McCarthy
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jessica M Conway
- Department of Mathematics, Pennsylvania State University, University Park, PA, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
4
|
Phan T, Conway JM, Pagane N, Kreig J, Sambaturu N, Iyaniwura S, Li JZ, Ribeiro RM, Ke R, Perelson AS. Understanding early HIV-1 rebound dynamics following antiretroviral therapy interruption: The importance of effector cell expansion. PLoS Pathog 2024; 20:e1012236. [PMID: 39074163 PMCID: PMC11309407 DOI: 10.1371/journal.ppat.1012236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/08/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Most people living with HIV-1 experience rapid viral rebound once antiretroviral therapy is interrupted; however, a small fraction remain in viral remission for an extended duration. Understanding the factors that determine whether viral rebound is likely after treatment interruption can enable the development of optimal treatment regimens and therapeutic interventions to potentially achieve a functional cure for HIV-1. We built upon the theoretical framework proposed by Conway and Perelson to construct dynamic models of virus-immune interactions to study factors that influence viral rebound dynamics. We evaluated these models using viral load data from 24 individuals following antiretroviral therapy interruption. The best-performing model accurately captures the heterogeneity of viral dynamics and highlights the importance of the effector cell expansion rate. Our results show that post-treatment controllers and non-controllers can be distinguished based on the effector cell expansion rate in our models. Furthermore, these results demonstrate the potential of using dynamic models incorporating an effector cell response to understand early viral rebound dynamics post-antiretroviral therapy interruption.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Jessica M. Conway
- Department of Mathematics, Pennsylvania State University, College Township, Pennsylvania, United States of America
- Department of Biology, Pennsylvania State University, College Township, Pennsylvania, United States of America
| | - Nicole Pagane
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, Massachusetts, United States of America
| | - Jasmine Kreig
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Narmada Sambaturu
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Sarafa Iyaniwura
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Ruian Ke
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| |
Collapse
|
5
|
Symmonds J, Gaufin T, Xu C, Raehtz KD, Ribeiro RM, Pandrea I, Apetrei C. Making a Monkey out of Human Immunodeficiency Virus/Simian Immunodeficiency Virus Pathogenesis: Immune Cell Depletion Experiments as a Tool to Understand the Immune Correlates of Protection and Pathogenicity in HIV Infection. Viruses 2024; 16:972. [PMID: 38932264 PMCID: PMC11209256 DOI: 10.3390/v16060972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Understanding the underlying mechanisms of HIV pathogenesis is critical for designing successful HIV vaccines and cure strategies. However, achieving this goal is complicated by the virus's direct interactions with immune cells, the induction of persistent reservoirs in the immune system cells, and multiple strategies developed by the virus for immune evasion. Meanwhile, HIV and SIV infections induce a pandysfunction of the immune cell populations, making it difficult to untangle the various concurrent mechanisms of HIV pathogenesis. Over the years, one of the most successful approaches for dissecting the immune correlates of protection in HIV/SIV infection has been the in vivo depletion of various immune cell populations and assessment of the impact of these depletions on the outcome of infection in non-human primate models. Here, we present a detailed analysis of the strategies and results of manipulating SIV pathogenesis through in vivo depletions of key immune cells populations. Although each of these methods has its limitations, they have all contributed to our understanding of key pathogenic pathways in HIV/SIV infection.
Collapse
Affiliation(s)
- Jen Symmonds
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Thaidra Gaufin
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433, USA;
| | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kevin D. Raehtz
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
6
|
Simpson J, Starke CE, Ortiz AM, Ransier A, Darko S, Llewellyn-Lacey S, Fennessey CM, Keele BF, Douek DC, Price DA, Brenchley JM. Immunotoxin-mediated depletion of Gag-specific CD8+ T cells undermines natural control of SIV. JCI Insight 2024; 9:e174168. [PMID: 38885329 PMCID: PMC11383179 DOI: 10.1172/jci.insight.174168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
Antibody-mediated depletion studies have demonstrated that CD8+ T cells are required for effective immune control of SIV. However, this approach is potentially confounded by several factors, including reactive CD4+ T cell proliferation, and provides no information on epitope specificity, a likely determinant of CD8+ T cell efficacy. We circumvented these limitations by selectively depleting CD8+ T cells specific for the Gag epitope CTPYDINQM (CM9) via the administration of immunotoxin-conjugated tetrameric complexes of CM9/Mamu-A*01. Immunotoxin administration effectively depleted circulating but not tissue-localized CM9-specific CD8+ T cells, akin to the bulk depletion pattern observed with antibodies directed against CD8. However, we found no evidence to indicate that circulating CM9-specific CD8+ T cells suppressed viral replication in Mamu-A*01+ rhesus macaques during acute or chronic progressive infection with a pathogenic strain of SIV. This observation extended to macaques with established infection during and after continuous antiretroviral therapy. In contrast, natural controller macaques experienced dramatic increases in plasma viremia after immunotoxin administration, highlighting the importance of CD8+ T cell-mediated immunity against CM9. Collectively, these data showed that CM9-specific CD8+ T cells were necessary but not sufficient for robust immune control of SIV in a nonhuman primate model and, more generally, validated an approach that could inform the design of next-generation vaccines against HIV-1.
Collapse
Affiliation(s)
- Jennifer Simpson
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Carly E Starke
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alexandra M Ortiz
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Amy Ransier
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sam Darko
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland, USA
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Sui Y, Meyer TJ, Fennessey CM, Keele BF, Dadkhah K, Ma C, LaBranche CC, Breed MW, Kramer JA, Li J, Howe SE, Ferrari G, Williams LD, Cam M, Kelly MC, Shen X, Tomaras GD, Montefiori D, Greten TF, Miller CJ, Berzofsky JA. Innate protection against intrarectal SIV acquisition by a live SHIV vaccine. JCI Insight 2024; 9:e175800. [PMID: 38912579 PMCID: PMC11383375 DOI: 10.1172/jci.insight.175800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/08/2024] [Indexed: 06/25/2024] Open
Abstract
Identifying immune correlates of protection is a major challenge in AIDS vaccine development. Anti-Envelope antibodies have been considered critical for protection against SIV/HIV (SHIV) acquisition. Here, we evaluated the efficacy of an SHIV vaccine against SIVmac251 challenge, where the role of antibody was excluded, as there was no cross-reactivity between SIV and SHIV envelope antibodies. After 8 low-dose intrarectal challenges with SIVmac251, 12 SHIV-vaccinated animals demonstrated efficacy, compared with 6 naive controls, suggesting protection was achieved in the absence of anti-envelope antibodies. Interestingly, CD8+ T cells (and some NK cells) were not essential for preventing viral acquisition, as none of the CD8-depleted macaques were infected by SIVmac251 challenges. Initial investigation of protective innate immunity revealed that protected animals had elevated pathways related to platelet aggregation/activation and reduced pathways related to interferon and responses to virus. Moreover, higher expression of platelet factor 4 on circulating platelet-leukocyte aggregates was associated with reduced viral acquisition. Our data highlighted the importance of innate immunity, identified mechanisms, and may provide opportunities for novel HIV vaccines or therapeutic strategy development.
Collapse
Affiliation(s)
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | | | | | - Kimia Dadkhah
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Chi Ma
- Thoracic and GI Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Celia C. LaBranche
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthew W. Breed
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Bethesda, Maryland, USA
| | - Josh A. Kramer
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Bethesda, Maryland, USA
| | | | | | | | - LaTonya D. Williams
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Michael C. Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - David Montefiori
- Duke Human Vaccine Institute and
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tim F. Greten
- Thoracic and GI Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Christopher J. Miller
- Center for Comparative Medicine, University of California, Davis, Davis, California, USA
| | | |
Collapse
|
8
|
Phan T, Conway JM, Pagane N, Kreig J, Sambaturu N, Iyaniwura S, Li JZ, Ribeiro RM, Ke R, Perelson AS. Understanding early HIV-1 rebound dynamics following antiretroviral therapy interruption: The importance of effector cell expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592318. [PMID: 38746144 PMCID: PMC11092759 DOI: 10.1101/2024.05.03.592318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Most people living with HIV-1 experience rapid viral rebound once antiretroviral therapy is interrupted; however, a small fraction remain in viral remission for an extended duration. Understanding the factors that determine whether viral rebound is likely after treatment interruption can enable the development of optimal treatment regimens and therapeutic interventions to potentially achieve a functional cure for HIV-1. We built upon the theoretical framework proposed by Conway and Perelson to construct dynamic models of virus-immune interactions to study factors that influence viral rebound dynamics. We evaluated these models using viral load data from 24 individuals following antiretroviral therapy interruption. The best-performing model accurately captures the heterogeneity of viral dynamics and highlights the importance of the effector cell expansion rate. Our results show that post-treatment controllers and non-controllers can be distinguished based on the effector cell expansion rate in our models. Furthermore, these results demonstrate the potential of using dynamic models incorporating an effector cell response to understand early viral rebound dynamics post-antiretroviral therapy interruption.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Jessica M Conway
- Department of Mathematics, Pennsylvania State University, College Township, PA, USA
- Department of Biology, Pennsylvania State University, College Township, PA, USA
| | - Nicole Pagane
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, USA
| | - Jasmine Kreig
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Narmada Sambaturu
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Sarafa Iyaniwura
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Jonathan Z Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Ruian Ke
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
- Santa Fe Institute, Santa Fe, NM, USA
| |
Collapse
|
9
|
Grasberger P, Sondrini AR, Clayton KL. Harnessing immune cells to eliminate HIV reservoirs. Curr Opin HIV AIDS 2024; 19:62-68. [PMID: 38167784 PMCID: PMC10908255 DOI: 10.1097/coh.0000000000000840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
PURPOSE OF REVIEW Despite decades of insights about how CD8 + T cells and natural killer (NK) cells contribute to natural control of infection, additional hurdles (mutational escape from cellular immunity, sequence diversity, and hard-to-access tissue reservoirs) will need to be overcome to develop a cure. In this review, we highlight recent findings of novel mechanisms of antiviral cellular immunity and discuss current strategies for therapeutic deisgn. RECENT FINDINGS Of note are the apparent converging roles of viral antigen-specific MHC-E-restricted CD8 + T cells and NK cells, interleukin (IL)-15 biologics to boost cytotoxicity, and broadly neutralizing antibodies in their native form or as anitbody fragments to neutralize virus and engage cellular immunity, respectively. Finally, renewed interest in myeloid cells as relevant viral reservoirs is an encouraging sign for designing inclusive therapeutic strategies. SUMMARY Several studies have shown promise in many preclinical models of disease, including simian immunodeficiency virus (SIV)/SHIV infection in nonhuman primates and HIV infection in humanized mice. However, each model comes with its own limitations and may not fully predict human responses. We eagerly await the results of clinical trails assessing the efficacy of these strategies to achieve reductions in viral reservoirs, delay viral rebound, or ultimately elicit immune based control of infection without combination antiretroviral therapy (cART).
Collapse
Affiliation(s)
- Paula Grasberger
- Department of Pathology, University of Massachusetts Chan Medical School
| | | | - Kiera L. Clayton
- Department of Pathology, University of Massachusetts Chan Medical School
| |
Collapse
|
10
|
Zeng YC, Young OJ, Si L, Ku MW, Isinelli G, Rajwar A, Jiang A, Wintersinger CM, Graveline AR, Vernet A, Sanchez M, Ryu JH, Kwon IC, Goyal G, Ingber DE, Shih WM. DNA origami vaccine (DoriVac) nanoparticles improve both humoral and cellular immune responses to infectious diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.29.573647. [PMID: 38260393 PMCID: PMC10802255 DOI: 10.1101/2023.12.29.573647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Current SARS-CoV-2 vaccines have demonstrated robust induction of neutralizing antibodies and CD4+ T cell activation, however CD8+ responses are variable, and the duration of immunity and protection against variants are limited. Here we repurposed our DNA origami vaccine platform, DoriVac, for targeting infectious viruses, namely SARS-CoV-2, HIV, and Ebola. The DNA origami nanoparticle, conjugated with infectious-disease-specific HR2 peptides, which act as highly conserved antigens, and CpG adjuvant at precise nanoscale spacing, induced neutralizing antibodies, Th1 CD4+ T cells, and CD8+ T cells in naïve mice, with significant improvement over a bolus control. Pre-clinical studies using lymph-node-on-a-chip systems validated that DoriVac, when conjugated with antigenic peptides or proteins, induced promising cellular immune responses in human cells. These results suggest that DoriVac holds potential as a versatile, modular vaccine platform, capable of inducing both humoral and cellular immunities. The programmability of this platform underscores its potential utility in addressing future pandemics.
Collapse
Affiliation(s)
- Yang C. Zeng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Olivia J. Young
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Longlong Si
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Min Wen Ku
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Giorgia Isinelli
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Anjali Rajwar
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Amanda Jiang
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Chris M. Wintersinger
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Amanda R. Graveline
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Andyna Vernet
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Melinda Sanchez
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Ju Hee Ryu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
| | - Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA
| | - William M. Shih
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
11
|
Cossarini F, Aberg JA, Chen BK, Mehandru S. Viral Persistence in the Gut-Associated Lymphoid Tissue and Barriers to HIV Cure. AIDS Res Hum Retroviruses 2023; 40:54-65. [PMID: 37450338 PMCID: PMC10790554 DOI: 10.1089/aid.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
More than 40 years after the first reported cases of what then became known as acquired immunodeficiency syndrome (AIDS), tremendous progress has been achieved in transforming the disease from almost universally fatal to a chronic manageable condition. Nonetheless, the efforts to find a preventative vaccine or a cure for the underlying infection with Human Immunodeficiency Virus (HIV) remain largely unsuccessful. Many challenges intrinsic to the virus characteristics and host response need to be overcome for either goal to be achieved. This article will review the obstacles to an effective HIV cure, specifically the steps involved in the generation of HIV latency, focusing on the role of the gut-associated lymphoid tissue, which has received less attention compared with the peripheral blood, despite being the largest repository of lymphoid tissue in the human body, and a large site for HIV persistence.
Collapse
Affiliation(s)
- Francesca Cossarini
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| | - Benjamin K. Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
- Division of Gastroenterology, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| |
Collapse
|
12
|
Evangelous TD, Berry M, Venkatayogi S, LeMaster C, Geanes ES, De Naeyer N, DeMarco T, Shen X, Li H, Hora B, Solomonis N, Misamore J, Lewis MG, Denny TN, Montefiori D, Shaw GM, Wiehe K, Bradley T, Williams WB. Host immunity associated with spontaneous suppression of viremia in therapy-naïve young rhesus macaques following neonatal SHIV infection. J Virol 2023; 97:e0109423. [PMID: 37874153 PMCID: PMC10688376 DOI: 10.1128/jvi.01094-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/06/2023] [Indexed: 10/25/2023] Open
Abstract
IMPORTANCE Despite the advent of highly active anti-retroviral therapy, people are still dying from HIV-related causes, many of whom are children, and a protective vaccine or cure is needed to end the HIV pandemic. Understanding the nature and activation states of immune cell subsets during infection will provide insights into the immunologic milieu associated with viremia suppression that can be harnessed via therapeutic strategies to achieve a functional cure, but these are understudied in pediatric subjects. We evaluated humoral and adaptive host immunity associated with suppression of viremia in rhesus macaques infected soon after birth with a pathogenic SHIV. The results from our study provide insights into the immune cell subsets and functions associated with viremia control in young macaques that may translate to pediatric subjects for the design of future anti-viral strategies in HIV-1-infected infants and children and contribute to an understudied area of HIV-1 pathogenesis in pediatric subjects.
Collapse
Affiliation(s)
- Tyler D. Evangelous
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sravani Venkatayogi
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Cas LeMaster
- Children’s Mercy Kansas City, Kansas City, Missouri, USA
| | - Eric S. Geanes
- Children’s Mercy Kansas City, Kansas City, Missouri, USA
| | - Nicole De Naeyer
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Todd DeMarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Surgery, Division of Surgical Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hui Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bhavna Hora
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | | | | - Thomas N. Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - David Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Surgery, Division of Surgical Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - George M. Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Todd Bradley
- Children’s Mercy Kansas City, Kansas City, Missouri, USA
- Department of Pediatrics, UMKC School of Medicine, Kansas City, Missouri, USA
- Departments of Pediatrics and Pathology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wilton B. Williams
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Surgery, Division of Surgical Sciences, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
13
|
Borgo GM, Rutishauser RL. Generating and measuring effective vaccine-elicited HIV-specific CD8 + T cell responses. Curr Opin HIV AIDS 2023; 18:331-341. [PMID: 37751362 PMCID: PMC10552829 DOI: 10.1097/coh.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW There is growing consensus that eliciting CD8 + T cells in addition to antibodies may be required for an effective HIV vaccine for both prevention and cure. Here, we review key qualities of vaccine-elicited CD8 + T cells as well as major CD8 + T cell-based delivery platforms used in recent HIV vaccine clinical trials. RECENT FINDINGS Much progress has been made in improving HIV immunogen design and delivery platforms to optimize CD8 + T cell responses. With regards to viral vectors, recent trials have tested newer chimp and human adenovirus vectors as well as a CMV vector. DNA vaccine immunogenicity has been increased by delivering the vaccines by electroporation and together with adjuvants as well as administering them as part of a heterologous regimen. In preclinical models, self-amplifying RNA vaccines can generate durable tissue-based CD8 + T cells. While it may be beneficial for HIV vaccines to recapitulate the functional and phenotypic features of HIV-specific CD8 + T cells isolated from elite controllers, most of these features are not routinely measured in HIV vaccine clinical trials. SUMMARY Identifying a vaccine capable of generating durable T cell responses that target mutationally vulnerable epitopes and that can rapidly intercept infecting or rebounding virus remains a challenge for HIV. Comprehensive assessment of HIV vaccine-elicited CD8 + T cells, as well as comparisons between different vaccine platforms, will be critical to advance our understanding of how to design better CD8 + T cell-based vaccines for HIV.
Collapse
Affiliation(s)
- Gina M Borgo
- Department of Medicine, University of California, San Francisco, California, USA
| | | |
Collapse
|
14
|
Policicchio BB, Cardozo-Ojeda EF, Xu C, Ma D, He T, Raehtz KD, Sivanandham R, Kleinman AJ, Perelson AS, Apetrei C, Pandrea I, Ribeiro RM. CD8 + T cells control SIV infection using both cytolytic effects and non-cytolytic suppression of virus production. Nat Commun 2023; 14:6657. [PMID: 37863982 PMCID: PMC10589330 DOI: 10.1038/s41467-023-42435-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/11/2023] [Indexed: 10/22/2023] Open
Abstract
Whether CD8+ T lymphocytes control human immunodeficiency virus infection by cytopathic or non-cytopathic mechanisms is not fully understood. Multiple studies highlighted non-cytopathic effects, but one hypothesis is that cytopathic effects of CD8+ T cells occur before viral production. Here, to examine the role of CD8+ T cells prior to virus production, we treated SIVmac251-infected macaques with an integrase inhibitor combined with a CD8-depleting antibody, or with either reagent alone. We analyzed the ensuing viral dynamics using a mathematical model that included infected cells pre- and post- viral DNA integration to compare different immune effector mechanisms. Macaques receiving the integrase inhibitor alone experienced greater viral load decays, reaching lower nadirs on treatment, than those treated also with the CD8-depleting antibody. Models including CD8+ cell-mediated reduction of viral production (non-cytolytic) were found to best explain the viral profiles across all macaques, in addition an effect in killing infected cells pre-integration (cytolytic) was supported in some of the best models. Our results suggest that CD8+ T cells have both a cytolytic effect on infected cells before viral integration, and a direct, non-cytolytic effect by suppressing viral production.
Collapse
Affiliation(s)
- Benjamin B Policicchio
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Dongzhu Ma
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Tianyu He
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Kevin D Raehtz
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ranjit Sivanandham
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Adam J Kleinman
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ivona Pandrea
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA.
- Laboratório de Biomatemática, Faculdade de Medicina da Universidade de Lisboa (previous address), Lisboa, Portugal.
| |
Collapse
|
15
|
Grant-McAuley W, Morgenlander W, Hudelson SE, Thakar M, Piwowar-Manning E, Clarke W, Breaud A, Blankson J, Wilson E, Ayles H, Bock P, Moore A, Kosloff B, Shanaube K, Meehan SA, van Deventer A, Fidler S, Hayes R, Ruczinski I, Kammers K, Laeyendecker O, Larman HB, Eshleman SH. Comprehensive profiling of pre-infection antibodies identifies HIV targets associated with viremic control and viral load. Front Immunol 2023; 14:1178520. [PMID: 37744365 PMCID: PMC10512082 DOI: 10.3389/fimmu.2023.1178520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023] Open
Abstract
Background High HIV viral load (VL) is associated with increased transmission risk and faster disease progression. HIV controllers achieve viral suppression without antiretroviral (ARV) treatment. We evaluated viremic control in a community-randomized trial with >48,000 participants. Methods A massively multiplexed antibody profiling system, VirScan, was used to quantify pre- and post-infection antibody reactivity to HIV peptides in 664 samples from 429 participants (13 controllers, 135 viremic non-controllers, 64 other non-controllers, 217 uninfected persons). Controllers had VLs <2,000 copies/mL with no ARV drugs detected at the first HIV-positive visit and one year later. Viremic non-controllers had VLs 2,000 copies/mL with no ARV drugs detected at the first HIV-positive visit. Other non-controllers had either ARV drugs detected at the first HIV-positive visit (n=47) or VLs <2,000 copies/mL with no ARV drugs detected at only one HIV-positive visit (n=17). Results We identified pre-infection HIV antibody reactivities that correlated with post-infection VL. Pre-infection reactivity to an epitope in the HR2 domain of gp41 was associated with controller status and lower VL. Pre-infection reactivity to an epitope in the C2 domain of gp120 was associated with non-controller status and higher VL. Different patterns of antibody reactivity were observed over time for these two epitopes. Conclusion These studies suggest that pre-infection HIV antibodies are associated with controller status and modulation of HIV VL. These findings may inform research on antibody-based interventions for HIV treatment.
Collapse
Affiliation(s)
- Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - William Morgenlander
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Manjusha Thakar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Estelle Piwowar-Manning
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - William Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Autumn Breaud
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joel Blankson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ethan Wilson
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Helen Ayles
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Bock
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | | | - Barry Kosloff
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kwame Shanaube
- Zambart, University of Zambia School of Public Health, Lusaka, Zambia
| | - Sue-Ann Meehan
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Anneen van Deventer
- Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Western Cape, South Africa
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Richard Hayes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kai Kammers
- Quantitative Sciences Division, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, MD, United States
| | - H. Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Nakamura-Hoshi M, Nomura T, Nishizawa M, Hau TTT, Yamamoto H, Okazaki M, Ishii H, Yonemitsu K, Suzaki Y, Ami Y, Matano T. HTLV-1 Proliferation after CD8 + Cell Depletion by Monoclonal Anti-CD8 Antibody Administration in Latently HTLV-1-Infected Cynomolgus Macaques. Microbiol Spectr 2023; 11:e0151823. [PMID: 37367230 PMCID: PMC10434050 DOI: 10.1128/spectrum.01518-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) induces chronic asymptomatic latent infection with a substantial proviral load but without significant viral replication in vivo. Cumulative studies have indicated involvement of CD8-positive (CD8+) cells, including virus-specific CD8+ T cells in the control of HTLV-1 replication. However, whether HTLV-1 expression from latently infected cells in vivo occurs in the absence of CD8+ cells remains unclear. Here, we examined the impact of CD8+ cell depletion by monoclonal anti-CD8 antibody administration on proviral load in HTLV-1-infected cynomolgus macaques. Five cynomolgus macaques were infected with HTLV-1 by inoculation with HTLV-1-producing cells. Administration of monoclonal anti-CD8 antibody in the chronic phase resulted in complete depletion of peripheral CD8+ T cells for approximately 2 months. All five macaques showed an increase in proviral load following CD8+ cell depletion, which peaked just before the reappearance of peripheral CD8+ T cells. Tax-specific CD8+ T-cell responses were detected in these recovered CD8+ T cells. Importantly, anti-HTLV-1 antibodies also increased after CD8+ cell depletion, indicating HTLV-1 antigen expression. These results provide evidence indicating that HTLV-1 can proliferate from the latent phase in the absence of CD8+ cells and suggest that CD8+ cells are responsible for the control of HTLV-1 replication. IMPORTANCE HTLV-1 can cause serious diseases such as adult T-cell leukemia (ATL) in humans after chronic asymptomatic latent infection with substantial proviral load. Proviruses are detectable in peripheral lymphocytes in HTLV-1 carriers, and the association of a higher proviral load with a higher risk of disease progression has been observed. However, neither substantial viral structural protein expression nor viral replication was detectable in vivo. Cumulative studies have indicated involvement of CD8+ cells, including virus-specific CD8+ T cells in the control of HTLV-1 replication. In the present study, we showed that CD8+ cell depletion by monoclonal anti-CD8 antibody administration results in HTLV-1 expression and an increase in proviral load in HTLV-1-infected cynomolgus macaques. Our results indicate that HTLV-1 can proliferate in the absence of CD8+ cells, suggesting that CD8+ cells are responsible for the control of HTLV-1 replication. This study provides insights into the mechanism of virus-host immune interaction in latent HTLV-1 infection.
Collapse
Affiliation(s)
| | - Takushi Nomura
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Masako Nishizawa
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Trang Thi Thu Hau
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroyuki Yamamoto
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Midori Okazaki
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroshi Ishii
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kenzo Yonemitsu
- Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuriko Suzaki
- Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yasushi Ami
- Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
De C, Pickles RJ, Yao W, Liao B, Boone A, Choi M, Battaglia DM, Askin FB, Whitmire JK, Silvestri G, Garcia JV, Wahl A. Human T cells efficiently control RSV infection. JCI Insight 2023; 8:e168110. [PMID: 37159271 PMCID: PMC10393221 DOI: 10.1172/jci.insight.168110] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection causes significant morbidity and mortality in infants, immunocompromised individuals, and older individuals. There is an urgent need for effective antivirals and vaccines for high-risk individuals. We used 2 complementary in vivo models to analyze RSV-associated human lung pathology and human immune correlates of protection. RSV infection resulted in widespread human lung epithelial damage, a proinflammatory innate immune response, and elicited a natural adaptive human immune response that conferred protective immunity. We demonstrated a key role for human T cells in controlling RSV infection. Specifically, primed human CD8+ T cells or CD4+ T cells effectively and independently control RSV replication in human lung tissue in the absence of an RSV-specific antibody response. These preclinical data support the development of RSV vaccines, which also elicit effective T cell responses to improve RSV vaccine efficacy.
Collapse
Affiliation(s)
- Chandrav De
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Raymond J. Pickles
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wenbo Yao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Baolin Liao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
- Department of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Allison Boone
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mingyu Choi
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Diana M. Battaglia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | | | - Jason K. Whitmire
- Department of Microbiology and Immunology, and
- Department of Genetics, and
- Lineberger Comprehensive Cancer Center, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - J. Victor Garcia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Angela Wahl
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| |
Collapse
|
18
|
Byrnes SJ, Busman-Sahay K, Angelovich TA, Younger S, Taylor-Brill S, Nekorchuk M, Bondoc S, Dannay R, Terry M, Cochrane CR, Jenkins TA, Roche M, Deleage C, Bosinger SE, Paiardini M, Brew BJ, Estes JD, Churchill MJ. Chronic immune activation and gut barrier dysfunction is associated with neuroinflammation in ART-suppressed SIV+ rhesus macaques. PLoS Pathog 2023; 19:e1011290. [PMID: 36989320 PMCID: PMC10085024 DOI: 10.1371/journal.ppat.1011290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 04/10/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
HIV-associated neurocognitive disorders (HAND) affect ~40% of virally suppressed people with HIV (PWH), however, the precise viral dependent and independent changes to the brain are unclear. Here we characterized the CNS reservoir and immune environment of SIV-infected (SIV+) rhesus macaques during acute (n = 4), chronic (n = 12) or ART-suppressed SIV infection (n = 11). Multiplex immunofluorescence for markers of SIV infection (vRNA/vDNA) and immune activation was performed on frontal cortex and matched colon tissue. SIV+ animals contained detectable viral DNA+ cells that were not reduced in the frontal cortex or the gut by ART, supporting the presence of a stable viral reservoir in these compartments. SIV+ animals had impaired blood brain barrier (BBB) integrity and heightened levels of astrocytes or myeloid cells expressing antiviral, anti-inflammatory or oxidative stress markers which were not abrogated by ART. Neuroinflammation and BBB dysfunction correlated with measures of viremia and immune activation in the gut. Furthermore, SIV-uninfected animals with experimentally induced gut damage and colitis showed a similar immune activation profile in the frontal cortex to those of SIV-infected animals, supporting the role of chronic gut damage as an independent source of neuroinflammation. Together, these findings implicate gut-associated immune activation/damage as a significant contributor to neuroinflammation in ART-suppressed HIV/SIV infection which may drive HAND pathogenesis.
Collapse
Affiliation(s)
- Sarah J. Byrnes
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Kathleen Busman-Sahay
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Thomas A. Angelovich
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
- Life Science, Burnet Institute, Melbourne, Australia
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Skyler Younger
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Sol Taylor-Brill
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Michael Nekorchuk
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Stephen Bondoc
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Rachel Dannay
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Margaret Terry
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | | | - Trisha A. Jenkins
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Michael Roche
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Bruce J. Brew
- Peter Duncan Neurosciences Unit, Departments of Neurology and Immunology St Vincent’s Hospital, University of New South Wales and University of Notre Dame, Sydney, New South Wales, Australia
| | - Jacob D. Estes
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Melissa J. Churchill
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
- Life Science, Burnet Institute, Melbourne, Australia
- Departments of Microbiology and Medicine, Monash University, Clayton, Australia
| |
Collapse
|
19
|
Statzu M, Jin W, Fray EJ, Wong AKH, Kumar MR, Ferrer E, Docken SS, Pinkevych M, McBrien JB, Fennessey CM, Keele BF, Liang S, Harper JL, Mutascio S, Franchitti L, Wang H, Cicetti D, Bosinger SE, Carnathan DG, Vanderford TH, Margolis DM, Garcia-Martinez JV, Chahroudi A, Paiardini M, Siliciano J, Davenport MP, Kulpa DA, Siliciano RS, Silvestri G. CD8 + lymphocytes do not impact SIV reservoir establishment under ART. Nat Microbiol 2023; 8:299-308. [PMID: 36690860 PMCID: PMC9894752 DOI: 10.1038/s41564-022-01311-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 12/15/2022] [Indexed: 01/24/2023]
Abstract
Persistence of the human immunodeficiency virus type-1 (HIV-1) latent reservoir in infected individuals remains a problem despite fully suppressive antiretroviral therapy (ART). While reservoir formation begins during acute infection, the mechanisms responsible for its establishment remain unclear. CD8+ T cells are important during the initial control of viral replication. Here we examined the effect of CD8+ T cells on formation of the latent reservoir in simian immunodeficiency virus (SIV)-infected macaques by performing experimental CD8+ depletion either before infection or before early (that is, day 14 post-infection) ART initiation. We found that CD8+ depletion resulted in slower decline of viremia, indicating that CD8+ lymphocytes reduce the average lifespan of productively infected cells during acute infection and early ART, presumably through SIV-specific cytotoxic T lymphocyte (CTL) activity. However, CD8+ depletion did not change the frequency of infected CD4+ T cells in the blood or lymph node as measured by the total cell-associated viral DNA or intact provirus DNA assay. In addition, the size of the persistent reservoir remained the same when measuring the kinetics of virus rebound after ART interruption. These data indicate that during early SIV infection, the viral reservoir that persists under ART is established largely independent of CTL control.
Collapse
Grants
- P30 AI050409 NIAID NIH HHS
- 75N91019D00024 NCI NIH HHS
- P51 OD011132 NIH HHS
- R01 AI143414 NIAID NIH HHS
- UM1 AI164562 NIAID NIH HHS
- UM1 AI164567 NIAID NIH HHS
- R01 AI125064 NIAID NIH HHS
- CU | National Cancer Institute, Cairo University (NCI)
- National Cancer Institute, National Institutes of Health, under Contract No. 75N91019D00024/HHSN261201500003I.
- This work was supported by UM1AI164562, co-funded by National Heart, Lung and Blood Institute, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Neurological Disorders and Stroke, National Institute on Drug Abuse and the National Institute of Allergy and Infectious Diseases (to G.S., D.A.K., M.P.1), and NIH NIAID R01-AI143414 (to G.S. and D.A.K), and R01-AI125064 (to G.S., A.C., D.A.K.).
Collapse
Affiliation(s)
- Maura Statzu
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Wang Jin
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Emily J Fray
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew Kam Ho Wong
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Mithra R Kumar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Ferrer
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steffen S Docken
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Mykola Pinkevych
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Julia B McBrien
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Shan Liang
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Justin L Harper
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Simona Mutascio
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Lavinia Franchitti
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Hong Wang
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Davide Cicetti
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Steven E Bosinger
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Diane G Carnathan
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Thomas H Vanderford
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - David M Margolis
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - J Victor Garcia-Martinez
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Ann Chahroudi
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Mirko Paiardini
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Janet Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Deanna A Kulpa
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Robert S Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guido Silvestri
- Emory National Primate Research Center, Department of Pathology and Laboratory Medicine, and Emory Vaccine Center, Emory University, Atlanta, GA, USA.
| |
Collapse
|
20
|
Gao L, Zhou J, Ye L. Role of CXCR5 + CD8 + T cells in human immunodeficiency virus-1 infection. Front Microbiol 2022; 13:998058. [PMID: 36452930 PMCID: PMC9701836 DOI: 10.3389/fmicb.2022.998058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection can be effectively suppressed by life-long administration of combination antiretroviral therapy (cART). However, the viral rebound can occur upon cART cessation due to the long-term presence of HIV reservoirs, posing a considerable barrier to drug-free viral remission. Memory CD4+ T cell subsets, especially T follicular helper (T FH ) cells that reside in B-cell follicles within lymphoid tissues, are regarded as the predominant cellular compartment of the HIV reservoir. Substantial evidence indicates that HIV-specific CD8+ T cell-mediated cellular immunity can sustain long-term disease-free and transmission-free HIV control in elite controllers. However, most HIV cure strategies that rely on expanded HIV-specific CD8+ T cells for virus control are likely to fail due to cellular exhaustion and T FH reservoir-specialized anatomical structures that isolate HIV-specific CD8+ T cell entry into B-cell follicles. Loss of stem-like memory properties is a key feature of exhaustion. Recent studies have found that CXC chemokine receptor type 5 (CXCR5)-expressing HIV-specific CD8+ T cells are memory-like CD8+ T cells that can migrate into B-cell follicles to execute inhibition of viral replication. Furthermore, these unique CD8+ T cells can respond to immune checkpoint blockade (ICB) therapy. In this review, we discuss the functions of these CD8+ T cells as well as the translation of findings into viable HIV treatment and cure strategies.
Collapse
Affiliation(s)
- Leiqiong Gao
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Zhou
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, China
| |
Collapse
|
21
|
Fardoos R, Nyquist SK, Asowata OE, Kazer SW, Singh A, Ngoepe A, Giandhari J, Mthabela N, Ramjit D, Singh S, Karim F, Buus S, Anderson F, Porterfield JZ, Sibiya AL, Bipath R, Moodley K, Kuhn W, Berger B, Nguyen S, de Oliveira T, Ndung’u T, Goulder P, Shalek AK, Leslie A, Kløverpris HN. HIV specific CD8 + T RM-like cells in tonsils express exhaustive signatures in the absence of natural HIV control. Front Immunol 2022; 13:912038. [PMID: 36330531 PMCID: PMC9623418 DOI: 10.3389/fimmu.2022.912038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Lymphoid tissues are an important HIV reservoir site that persists in the face of antiretroviral therapy and natural immunity. Targeting these reservoirs by harnessing the antiviral activity of local tissue-resident memory (TRM) CD8+ T-cells is of great interest, but limited data exist on TRM-like cells within lymph nodes of people living with HIV (PLWH). Here, we studied tonsil CD8+ T-cells obtained from PLWH and uninfected controls from South Africa. We show that these cells are preferentially located outside the germinal centers (GCs), the main reservoir site for HIV, and display a low cytolytic and a transcriptionally TRM-like profile distinct from blood CD8+ T-cells. In PLWH, CD8+ TRM-like cells are expanded and adopt a more cytolytic, activated, and exhausted phenotype not reversed by antiretroviral therapy (ART). This phenotype was enhanced in HIV-specific CD8+ T-cells from tonsils compared to matched blood suggesting a higher antigen burden in tonsils. Single-cell transcriptional and clonotype resolution showed that these HIV-specific CD8+ T-cells in the tonsils express heterogeneous signatures of T-cell activation, clonal expansion, and exhaustion ex-vivo. Interestingly, this signature was absent in a natural HIV controller, who expressed lower PD-1 and CXCR5 levels and reduced transcriptional evidence of T-cell activation, exhaustion, and cytolytic activity. These data provide important insights into lymphoid tissue-derived HIV-specific CD8+ TRM-like phenotypes in settings of HIV remission and highlight their potential for immunotherapy and targeting of the HIV reservoirs.
Collapse
Affiliation(s)
- Rabiah Fardoos
- Africa Health Research Institute (AHRI), Durban, South Africa
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sarah K. Nyquist
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | - Samuel W. Kazer
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Alveera Singh
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Abigail Ngoepe
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Jennifer Giandhari
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Dirhona Ramjit
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Samita Singh
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Farina Karim
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Frank Anderson
- Discipline of General Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - J. Zachary Porterfield
- Africa Health Research Institute (AHRI), Durban, South Africa
- Department of Otolaryngology-Head & Neck Surgery, Division of Infectious Diseases, University of Kentucky, Lexington, KY, United States
- Department of Microbiology, Immunology and Molecular Genetics, - Division of Infectious Diseases, University of Kentucky, Lexington, KY, United States
- Department of Internal Medicine - Division of Infectious Diseases, University of Kentucky, Lexington, KY, United States
| | - Andile L. Sibiya
- Department of Otorhinolaryngology & Head & Neck Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Rishan Bipath
- Department of Otorhinolaryngology, King Edward VIII hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Kumeshan Moodley
- Department of Ear Nose and Throat, General Justice Gizenga Mpanza Regional Hospital (Stanger Hospital), University of KwaZulu-Natal, Durban, South Africa
| | - Warren Kuhn
- Department of Otorhinolaryngology & Head & Neck Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
- Department of Ear Nose and Throat, General Justice Gizenga Mpanza Regional Hospital (Stanger Hospital), University of KwaZulu-Natal, Durban, South Africa
| | - Bonnie Berger
- Computer Science & Artificial Intelligence Lab and Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Son Nguyen
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Tulio de Oliveira
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thumbi Ndung’u
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa
- University College London, Division of Infection and Immunity, London, United Kingdom
| | - Philip Goulder
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Alex K. Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, United States
- Ragon Institute of MGH, Harvard, Cambridge, MA, United States
| | - Alasdair Leslie
- Africa Health Research Institute (AHRI), Durban, South Africa
- University College London, Division of Infection and Immunity, London, United Kingdom
| | - Henrik N. Kløverpris
- Africa Health Research Institute (AHRI), Durban, South Africa
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- University College London, Division of Infection and Immunity, London, United Kingdom
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Immunological studies of spontaneous HIV and simian virus (SIV) controllers have identified virus-specific CD8 + T cells as a key immune mechanism of viral control. The purpose of this review is to consider how knowledge about the mechanisms that are associated with CD8 + T cell control of HIV/SIV in natural infection can be harnessed in HIV remission strategies. RECENT FINDINGS We discuss characteristics of CD8 + T-cell responses that may be critical for suppressing HIV replication in spontaneous controllers comprising HIV antigen recognition including specific human leukocyte antigen types, broadly cross-reactive T cell receptors and epitope targeting, enhanced expansion and antiviral functions, and localization of virus-specific T cells near sites of reservoir persistence. We also discuss the need to better understand the timing of CD8 + T-cell responses associated with viral control of HIV/SIV during acute infection and after treatment interruption as well as the mechanisms by which HIV/SIV-specific CD8 + T cells coordinate with other immune responses to achieve control. SUMMARY We propose implications as to how this knowledge from natural infection can be applied in the design and evaluation of CD8 + T-cell-based remission strategies and offer questions to consider as these strategies target distinct CD8 + T-cell-dependent mechanisms of viral control.
Collapse
|
23
|
Zheng HY, Wang XH, He XY, Chen M, Zhang MX, Lian XD, Song JH, Hu Y, Pang W, Wang Y, Hu ZF, Lv LB, Zheng YT. Aging induces severe SIV infection accompanied by an increase in follicular CD8+ T cells with overactive STAT3 signaling. Cell Mol Immunol 2022; 19:1042-1053. [PMID: 35851876 PMCID: PMC9424273 DOI: 10.1038/s41423-022-00899-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
The number of elderly people living with HIV is increasing globally, and the condition of this population is relatively complicated due to the dual effects of aging and HIV infection. However, the impact of HIV infection combined with aging on the immune homeostasis of secondary lymphoid organs remains unclear. Here, we used the simian immunodeficiency virus mac239 (SIVmac239) strain to infect six young and six old Chinese rhesus macaques (ChRMs) and compared the infection characteristics of the two groups in the chronic stage through multiplex immunofluorescence staining of lymph nodes. The results showed that the SIV production and CD4/CD8 ratio inversion in old ChRMs were more severe than those in young ChRMs in both the peripheral blood and the lymph nodes, especially when a large number of CD8+ T cells infiltrated the follicles and germinal centers. STAT3 in these follicular CXCR5+CD8+ T cells was highly activated, with high expression of granzyme B, which might be caused by the severe inflammatory milieu in the follicles of old ChRMs. This study indicates that aging may be a cofactor involved in SIV-induced immune disorders in secondary lymphoid tissues, affecting the effective antiviral activity of highly enriched follicular CXCR5+CD8+ cells.
Collapse
Affiliation(s)
- Hong-Yi Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Xue-Hui Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xiao-Yan He
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Min Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ming-Xu Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Xiao-Dong Lian
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Jia-Hao Song
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yan Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Wei Pang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Yun Wang
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Zheng-Fei Hu
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Long-Bao Lv
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230026, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China.
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| |
Collapse
|
24
|
Berendam SJ, Nelson AN, Yagnik B, Goswami R, Styles TM, Neja MA, Phan CT, Dankwa S, Byrd AU, Garrido C, Amara RR, Chahroudi A, Permar SR, Fouda GG. Challenges and Opportunities of Therapies Targeting Early Life Immunity for Pediatric HIV Cure. Front Immunol 2022; 13:885272. [PMID: 35911681 PMCID: PMC9325996 DOI: 10.3389/fimmu.2022.885272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
Early initiation of antiretroviral therapy (ART) significantly improves clinical outcomes and reduces mortality of infants/children living with HIV. However, the ability of infected cells to establish latent viral reservoirs shortly after infection and to persist during long-term ART remains a major barrier to cure. In addition, while early ART treatment of infants living with HIV can limit the size of the virus reservoir, it can also blunt HIV-specific immune responses and does not mediate clearance of latently infected viral reservoirs. Thus, adjunctive immune-based therapies that are geared towards limiting the establishment of the virus reservoir and/or mediating the clearance of persistent reservoirs are of interest for their potential to achieve viral remission in the setting of pediatric HIV. Because of the differences between the early life and adult immune systems, these interventions may need to be tailored to the pediatric settings. Understanding the attributes and specificities of the early life immune milieu that are likely to impact the virus reservoir is important to guide the development of pediatric-specific immune-based interventions towards viral remission and cure. In this review, we compare the immune profiles of pediatric and adult HIV elite controllers, discuss the characteristics of cellular and anatomic HIV reservoirs in pediatric populations, and highlight the potential values of current cure strategies using immune-based therapies for long-term viral remission in the absence of ART in children living with HIV.
Collapse
Affiliation(s)
- Stella J. Berendam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States,*Correspondence: Stella J. Berendam, ; Genevieve G. Fouda,
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Bhrugu Yagnik
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Ria Goswami
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Tiffany M. Styles
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Margaret A. Neja
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Caroline T. Phan
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Sedem Dankwa
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Alliyah U. Byrd
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Carolina Garrido
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Rama R. Amara
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States,Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States,*Correspondence: Stella J. Berendam, ; Genevieve G. Fouda,
| |
Collapse
|
25
|
Kumar Singh A, Padwal V, Palav H, Velhal S, Nagar V, Patil P, Patel V. Highly dampened HIV-specific cytolytic effector T cell responses define viremic non-progression. Immunobiology 2022; 227:152234. [PMID: 35671626 DOI: 10.1016/j.imbio.2022.152234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/30/2022] [Indexed: 11/05/2022]
Abstract
This study reports on HIV-specific T cell responses in HIV-1 infected Viremic Non-Progressors (VNPs), a rare group of people living with HIV that exhibit asymptomatic infection over several years accompanied by stable CD4+ T cell counts in spite of ongoing viral replication. We attempted to identify key virus-specific functional attributes that could underlie the apparently paradoxical virus-host equilibrium observed in VNPs. Our results revealed modulation of HIV-specific CD4+ and CD8+ effector T cell responses in VNPs towards a dominant non-cytolytic profile with concomitantly diminished degranulation (CD107a+) ability. Further, the HIV specific CD8+ effector T cell response was primarily enriched for MIP-1β producing cells. As expected, concordant with better viral suppression, VCs exhibit a robust cytolytic T cell response. Interestingly, PuPs shared features common to both these responses but did not exhibit a CD4+ central memory IFN-γ producing Gag-specific response that was shared by both non-progressor (VC and VNP) groups, suggesting CD4 helper response is critical for non-progression. Our study also revealed that cytolytic response in VNPs is primarily limited to polyfunctional cells while both monofunctional and polyfunctional cells significantly contribute to cytolytic responses in VCs. To further understand mechanisms underlying the unique HIV-specific effector T cell response described here in VNPs we also evaluated and demonstrated a possible role for altered gut homing in these individuals. Our findings inform immunotherapeutic interventions to achieve functional cures in the context of ART resistance and serious non AIDS events.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Varsha Padwal
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Harsha Palav
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Shilpa Velhal
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Vidya Nagar
- Department of Medicine, Grant Medical College & Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
| | - Priya Patil
- Department of Medicine, Grant Medical College & Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
| | - Vainav Patel
- Viral Immunopathogenesis Laboratory, ICMR-National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India.
| |
Collapse
|
26
|
Collins DR, Urbach JM, Racenet ZJ, Arshad U, Power KA, Newman RM, Mylvaganam GH, Ly NL, Lian X, Rull A, Rassadkina Y, Yanez AG, Peluso MJ, Deeks SG, Vidal F, Lichterfeld M, Yu XG, Gaiha GD, Allen TM, Walker BD. Functional impairment of HIV-specific CD8 + T cells precedes aborted spontaneous control of viremia. Immunity 2021; 54:2372-2384.e7. [PMID: 34496223 PMCID: PMC8516715 DOI: 10.1016/j.immuni.2021.08.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/21/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
Spontaneous control of HIV infection has been repeatedly linked to antiviral CD8+ T cells but is not always permanent. To address mechanisms of durable and aborted control of viremia, we evaluated immunologic and virologic parameters longitudinally among 34 HIV-infected subjects with differential outcomes. Despite sustained recognition of autologous virus, HIV-specific proliferative and cytolytic T cell effector functions became selectively and intrinsically impaired prior to aborted control. Longitudinal transcriptomic profiling of functionally impaired HIV-specific CD8+ T cells revealed altered expression of genes related to activation, cytokine-mediated signaling, and cell cycle regulation, including increased expression of the antiproliferative transcription factor KLF2 but not of genes associated with canonical exhaustion. Lymphoid HIV-specific CD8+ T cells also exhibited poor functionality during aborted control relative to durable control. Our results identify selective functional impairment of HIV-specific CD8+ T cells as prognostic of impending aborted HIV control, with implications for clinical monitoring and immunotherapeutic strategies.
Collapse
Affiliation(s)
- David R Collins
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | | | - Umar Arshad
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Karen A Power
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Ruchi M Newman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Geetha H Mylvaganam
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Ngoc L Ly
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Xiaodong Lian
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Anna Rull
- Joan XXIII University Hospital, Pere Virgili Institute (IISPV), Rovira i Virgili University, Tarragona, Spain
| | - Yelizaveta Rassadkina
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Adrienne G Yanez
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Michael J Peluso
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, USA
| | - Francesc Vidal
- Joan XXIII University Hospital, Pere Virgili Institute (IISPV), Rovira i Virgili University, Tarragona, Spain
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Xu G Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Gaurav D Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Institute for Medical Engineering and Sciences and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
27
|
Zhou Y, Maldini CR, Jadlowsky J, Riley JL. Challenges and Opportunities of Using Adoptive T-Cell Therapy as Part of an HIV Cure Strategy. J Infect Dis 2021; 223:38-45. [PMID: 33586770 DOI: 10.1093/infdis/jiaa223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
HIV-infected individuals successfully controlling viral replication via antiretroviral therapy often have a compromised HIV-specific T-cell immune response due to the lack of CD4 T-cell help, viral escape, T-cell exhaustion, and reduction in numbers due to the withdrawal of cognate antigen. A successful HIV cure strategy will likely involve a durable and potent police force that can effectively recognize and eliminate remaining virus that may emerge decades after an individual undergoes an HIV cure regimen. T cells are ideally suited to serve in this role, but given the state of the HIV-specific T-cell response, it is unclear how to best restore HIV-specific T-cell activity prior initiation of a HIV cure strategy. Here, we review several strategies of generating HIV-specific T cells ex vivo that are currently being tested in the clinic and discuss how infused T cells can be part of an HIV cure strategy.
Collapse
Affiliation(s)
- Yuqi Zhou
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Colby R Maldini
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Julie Jadlowsky
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James L Riley
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Bricker KM, Chahroudi A, Mavigner M. New Latency Reversing Agents for HIV-1 Cure: Insights from Nonhuman Primate Models. Viruses 2021; 13:1560. [PMID: 34452425 PMCID: PMC8402914 DOI: 10.3390/v13081560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/26/2021] [Accepted: 08/03/2021] [Indexed: 01/30/2023] Open
Abstract
Antiretroviral therapy (ART) controls human immunodeficiency virus 1 (HIV-1) replication and prevents disease progression but does not eradicate HIV-1. The persistence of a reservoir of latently infected cells represents the main barrier to a cure. "Shock and kill" is a promising strategy involving latency reversing agents (LRAs) to reactivate HIV-1 from latently infected cells, thus exposing the infected cells to killing by the immune system or clearance agents. Here, we review advances to the "shock and kill" strategy made through the nonhuman primate (NHP) model, highlighting recently identified latency reversing agents and approaches such as mimetics of the second mitochondrial activator of caspase (SMACm), experimental CD8+ T cell depletion, immune checkpoint blockade (ICI), and toll-like receptor (TLR) agonists. We also discuss the advantages and limits of the NHP model for HIV cure research and methods developed to evaluate the efficacy of in vivo treatment with LRAs in NHPs.
Collapse
Affiliation(s)
- Katherine M. Bricker
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (K.M.B.); (A.C.)
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (K.M.B.); (A.C.)
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory + Children’s Center for Childhood Infections and Vaccines, Atlanta, GA 30322, USA
| | - Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (K.M.B.); (A.C.)
| |
Collapse
|
29
|
Potential Utility of Natural Killer Cells for Eliminating Cells Harboring Reactivated Latent HIV-1 Following the Removal of CD8 + T Cell-Mediated Pro-Latency Effect(s). Viruses 2021; 13:v13081451. [PMID: 34452317 PMCID: PMC8402732 DOI: 10.3390/v13081451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
An impediment to curing HIV-1 infection is the persistence of latently infected cells in ART-treated people living with HIV (PLWH). A key strategy for curing HIV-1 infection is to activate transcription and translation of latent virus using latency reversing agents (LRAs) and eliminate cells harboring reactivated virus via viral cytopathic effect or immune clearance. In this review, we provide an overview of available LRAs and their use in clinical trials. Furthermore, we describe recent data suggesting that CD8+ T cells promote HIV-1 latency in the context of ART, even in the presence of LRAs, which might at least partially explain the clinical inefficiency of previous “shock and kill” trials. Here, we propose a novel cure strategy called “unlock, shock, disarm, and kill”. The general premise of this strategy is to shut down the pro-latency function(s) of CD8+ T cells, use LRAs to reverse HIV-1 latency, counteract anti-apoptotic molecules, and engage natural killer (NK) cells to mediate the killing of cells harboring reactivated latent HIV-1.
Collapse
|
30
|
Kula-Pacurar A, Rodari A, Darcis G, Van Lint C. Shocking HIV-1 with immunomodulatory latency reversing agents. Semin Immunol 2021; 51:101478. [PMID: 33972164 DOI: 10.1016/j.smim.2021.101478] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
The "shock-and-kill" strategy is one of the most explored HIV-1 cure approaches to eliminate latent virus. This strategy is based on HIV-1 reactivation using latency reversing agents (LRAs) to reactivate latent proviruses (the "shock" phase) and to induce subsequent elimination of the reactivated cells by immune responses or virus-induced cytopathic effects (the "kill" phase). Studies using immunomodulatory LRAs such as blockers of immune checkpoint molecules, toll-like receptor agonists, cytokines and CD8+ T cell depleting antibodies showed promising potential as LRAs inducing directly or indirectly cellular pathways known to control HIV transcription. However, the precise molecular mechanisms by which these immunomodulatory LRAs reverse latency remain incompletely understood. Together with the heterogenous nature of HIV-1 latency, this lack of understanding complicates efforts to develop more efficient and safer cure strategies. Hence, deciphering those mechanisms is pivotal in designing approaches to eliminate latent HIV infection.
Collapse
Affiliation(s)
- Anna Kula-Pacurar
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Anthony Rodari
- Service of Molecular Virology, Department of Molecular Virology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Gilles Darcis
- Infectious Diseases Department, Liège University Hospital, Liège, Belgium
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Virology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium.
| |
Collapse
|
31
|
Optimal Maturation of the SIV-Specific CD8 + T Cell Response after Primary Infection Is Associated with Natural Control of SIV: ANRS SIC Study. Cell Rep 2021; 32:108174. [PMID: 32966788 DOI: 10.1016/j.celrep.2020.108174] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 07/10/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
Highly efficient CD8+ T cells are associated with natural HIV control, but it has remained unclear how these cells are generated and maintained. We have used a macaque model of spontaneous SIVmac251 control to monitor the development of efficient CD8+ T cell responses. Our results show that SIV-specific CD8+ T cells emerge during primary infection in all animals. The ability of CD8+ T cells to suppress SIV is suboptimal in the acute phase but increases progressively in controller macaques before the establishment of sustained low-level viremia. Controller macaques develop optimal memory-like SIV-specific CD8+ T cells early after infection. In contrast, a persistently skewed differentiation phenotype characterizes memory SIV-specific CD8+ T cells in non-controller macaques. Accordingly, the phenotype of SIV-specific CD8+ T cells defined early after infection appears to favor the development of protective immunity in controllers, whereas SIV-specific CD8+ T cells in non-controllers fail to gain antiviral potency, feasibly as a consequence of early defects imprinted in the memory pool.
Collapse
|
32
|
Assis MA, Carranza PG, Ambrosio E. A "Drug-Dependent" Immune System Can Compromise Protection against Infection: The Relationships between Psychostimulants and HIV. Viruses 2021; 13:v13050722. [PMID: 33919273 PMCID: PMC8143316 DOI: 10.3390/v13050722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/31/2023] Open
Abstract
Psychostimulant use is a major comorbidity in people living with HIV, which was initially explained by them adopting risky behaviors that facilitate HIV transmission. However, the effects of drug use on the immune system might also influence this phenomenon. Psychostimulants act on peripheral immune cells even before they reach the central nervous system (CNS) and their effects on immunity are likely to influence HIV infection. Beyond their canonical activities, classic neurotransmitters and neuromodulators are expressed by peripheral immune cells (e.g., dopamine and enkephalins), which display immunomodulatory properties and could be influenced by psychostimulants. Immune receptors, like Toll-like receptors (TLRs) on microglia, are modulated by cocaine and amphetamine exposure. Since peripheral immunocytes also express TLRs, they may be similarly affected by psychostimulants. In this review, we will summarize how psychostimulants are currently thought to influence peripheral immunity, mainly focusing on catecholamines, enkephalins and TLR4, and shed light on how these drugs might affect HIV infection. We will try to shift from the classic CNS perspective and adopt a more holistic view, addressing the potential impact of psychostimulants on the peripheral immune system and how their systemic effects could influence HIV infection.
Collapse
Affiliation(s)
- María Amparo Assis
- Facultad de Ciencias Médicas, Universidad Nacional de Santiago del Estero (UNSE), Santiago del Estero G4200, Argentina;
- Laboratorio de Biología Molecular, Inmunología y Microbiología, Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero G4206, Argentina
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
- Correspondence:
| | - Pedro Gabriel Carranza
- Facultad de Ciencias Médicas, Universidad Nacional de Santiago del Estero (UNSE), Santiago del Estero G4200, Argentina;
- Laboratorio de Biología Molecular, Inmunología y Microbiología, Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD), CONICET-UNSE, Santiago del Estero G4206, Argentina
- Facultad de Agronomía y Agroindustrias, Universidad Nacional de Santiago del Estero, Santiago del Estero G4206, Argentina
| | - Emilio Ambrosio
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
| |
Collapse
|
33
|
CD8 lymphocyte depletion enhances the latency reversal activity of the SMAC mimetic AZD5582 in ART-suppressed SIV-infected rhesus macaques. J Virol 2021; 95:JVI.01429-20. [PMID: 33568515 PMCID: PMC8103677 DOI: 10.1128/jvi.01429-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inducing latency reversal to reveal infected cells to the host immune system represents a potential strategy to cure HIV infection. In separate studies, we have previously shown that CD8+ T cells may contribute to the maintenance of viral latency and identified a novel SMAC mimetic/IAP inhibitor (AZD5582) capable of reversing HIV/SIV latency in vivo by activating the non-canonical (nc) NF-κB pathway. Here, we use AZD5582 in combination with antibody-mediated depletion of CD8α+ cells to further evaluate the role of CD8+ T cells in viral latency maintenance. Six rhesus macaques (RM) were infected with SIVmac239 and treated with ART starting at week 8 post-infection. After 84-85 weeks of ART, all animals received a single dose of the anti-CD8α depleting antibody (Ab), MT807R1 (50mg/kg, s.c.), followed by 5 weekly doses of AZD5582 (0.1 mg/kg, i.v.). Following CD8α depletion + AZD5582 combined treatment, 100% of RMs experienced on-ART viremia above 60 copies per ml of plasma. In comparator groups of ART-suppressed SIV-infected RMs treated with AZD5582 only or CD8α depletion only, on-ART viremia was experienced by 56% and 57% of the animals respectively. Furthermore, the frequency of increased viremic episodes during the treatment period was greater in the CD8α depletion + AZD5582 group as compared to other groups. Mathematical modeling of virus reactivation suggested that, in addition to viral dynamics during acute infection, CD8α depletion influenced the response to AZD5582. This work suggests that the latency reversal induced by activation of the ncNF-κB signaling pathway with AZD5582 can be enhanced by CD8α+ cell depletion.
Collapse
|
34
|
Immunologic Control of HIV-1: What Have We Learned and Can We Induce It? Curr HIV/AIDS Rep 2021; 18:211-220. [PMID: 33709324 DOI: 10.1007/s11904-021-00545-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW A large amount of data now exists on the virus-specific immune response associated with spontaneous or induced immunologic control of lentiviruses. This review focuses on how the current understanding of HIV-specific immunity might be leveraged into induction of immunologic control and what further research is needed to accomplish this goal. RECENT FINDINGS During chronic infection, the function most robustly associated with immunologic control of HIV-1 is CD8+ T cell cytotoxic capacity. This function has proven difficult to restore in HIV-specific CD8+ T cells of chronically infected progressors in vitro and in vivo. However, progress has been made in inducing an effective CD8+ T cell response prior to lentiviral infection in the macaque model and during acute lentiviral infection in non-human primates. Further study will likely accelerate the ability to induce an effective CD8+ T cell response as part of prophylactic or therapeutic strategies.
Collapse
|
35
|
Rutishauser RL, Deguit CDT, Hiatt J, Blaeschke F, Roth TL, Wang L, Raymond KA, Starke CE, Mudd JC, Chen W, Smullin C, Matus-Nicodemos R, Hoh R, Krone M, Hecht FM, Pilcher CD, Martin JN, Koup RA, Douek DC, Brenchley JM, Sékaly RP, Pillai SK, Marson A, Deeks SG, McCune JM, Hunt PW. TCF-1 regulates HIV-specific CD8+ T cell expansion capacity. JCI Insight 2021; 6:136648. [PMID: 33351785 PMCID: PMC7934879 DOI: 10.1172/jci.insight.136648] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Although many HIV cure strategies seek to expand HIV-specific CD8+ T cells to control the virus, all are likely to fail if cellular exhaustion is not prevented. A loss in stem-like memory properties (i.e., the ability to proliferate and generate secondary effector cells) is a key feature of exhaustion; little is known, however, about how these properties are regulated in human virus-specific CD8+ T cells. We found that virus-specific CD8+ T cells from humans and nonhuman primates naturally controlling HIV/SIV infection express more of the transcription factor TCF-1 than noncontrollers. HIV-specific CD8+ T cell TCF-1 expression correlated with memory marker expression and expansion capacity and declined with antigenic stimulation. CRISPR-Cas9 editing of TCF-1 in human primary T cells demonstrated a direct role in regulating expansion capacity. Collectively, these data suggest that TCF-1 contributes to the regulation of the stem-like memory property of secondary expansion capacity of HIV-specific CD8+ T cells, and they provide a rationale for exploring the enhancement of this pathway in T cell-based therapeutic strategies for HIV.
Collapse
Affiliation(s)
| | - Christian Deo T. Deguit
- Department of Medicine, UCSF, San Francisco, California, USA
- Institute of Human Genetics, University of the Philippines-National Institutes of Health, Manila, Philippines
| | - Joseph Hiatt
- Department of Microbiology and Immunology
- Medical Scientist Training Program
- Biomedical Sciences Graduate Program, and
| | - Franziska Blaeschke
- Department of Microbiology and Immunology
- Diabetes Center, UCSF, San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, USA
| | - Theodore L. Roth
- Department of Microbiology and Immunology
- Medical Scientist Training Program
- Biomedical Sciences Graduate Program, and
| | - Lynn Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Kyle A. Raymond
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, California, USA
| | | | - Joseph C. Mudd
- Barrier Immunity Section, Laboratory of Viral Diseases and
| | - Wenxuan Chen
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Carolyn Smullin
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Rodrigo Matus-Nicodemos
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rebecca Hoh
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Melissa Krone
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California, USA
| | | | | | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California, USA
| | - Richard A. Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases , NIH, Bethesda, Maryland, USA
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | | | - Satish K. Pillai
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, California, USA
| | - Alexander Marson
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Microbiology and Immunology
- Diabetes Center, UCSF, San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
- UCSF Hellen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Steven G. Deeks
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Peter W. Hunt
- Department of Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
36
|
Zanoni M, Palesch D, Pinacchio C, Statzu M, Tharp GK, Paiardini M, Chahroudi A, Bosinger SE, Yoon J, Cox B, Silvestri G, Kulpa DA. Innate, non-cytolytic CD8+ T cell-mediated suppression of HIV replication by MHC-independent inhibition of virus transcription. PLoS Pathog 2020; 16:e1008821. [PMID: 32941545 PMCID: PMC7523993 DOI: 10.1371/journal.ppat.1008821] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 09/29/2020] [Accepted: 07/18/2020] [Indexed: 12/31/2022] Open
Abstract
MHC-I-restricted, virus-specific cytotoxic CD8+ T cells (CTLs) may control human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replication via the recognition and killing of productively infected CD4+ T cells. Several studies in SIV-infected macaques suggest that CD8+ T cells may also decrease virus production by suppressing viral transcription. Here, we show that non-HIV-specific, TCR-activated non-cytolytic CD8+ T cells suppress HIV transcription via a virus- and MHC-independent immunoregulatory mechanism that modulates CD4+ T cell proliferation and activation. We also demonstrate that this CD8+ T cell-mediated effect promotes the survival of infected CD4+ T cells harboring integrated, inducible virus. Finally, we used RNA sequencing and secretome analyses to identify candidate cellular pathways that are involved in the virus-silencing mediated by these CD8+ T cells. This study characterizes a previously undescribed mechanism of immune-mediated HIV silencing that may be involved in the establishment and maintenance of the reservoir under antiretroviral therapy and therefore represent a major obstacle to HIV eradication.
Collapse
Affiliation(s)
- Michelle Zanoni
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - David Palesch
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - Claudia Pinacchio
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - Maura Statzu
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - Gregory K. Tharp
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
| | - Jack Yoon
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Bryan Cox
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Deanna A. Kulpa
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Emory Vaccine Center Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
37
|
Therapeutic vaccination with IDLV-SIV-Gag results in durable viremia control in chronically SHIV-infected macaques. NPJ Vaccines 2020; 5:36. [PMID: 32411399 PMCID: PMC7210278 DOI: 10.1038/s41541-020-0186-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/17/2020] [Indexed: 01/14/2023] Open
Abstract
Despite incredible scientific efforts, there is no cure for HIV infection. While antiretroviral treatment (ART) can help control the virus and prevent transmission, it cannot eradicate HIV from viral reservoirs established before the initiation of therapy. Further, HIV-infected individuals reliably exhibit viral rebound when ART is interrupted, suggesting that the host immune response fails to control viral replication in persistent reservoirs. Therapeutic vaccines are one current approach to improving antiviral host immune responses and enhance long term virus control. In the present study, we used an integrase defective lentiviral vector (IDLV) expressing SIV-Gag to boost anti-Gag specific immune responses in macaques chronically infected with the tier-2 SHIV-1157(QNE)Y173H. A single immunization with IDLV-SIV-Gag induced durable (>20 weeks) virus control in 55% of the vaccinated macaques, correlating with an increased magnitude of SIV-Gag specific CD8+ T-cell responses. IDLV-based therapeutic vaccines are therefore an effective approach to improve virus specific CD8+ T-cell responses and mediate virus control.
Collapse
|
38
|
Penkert RR, Hankins JS, Young NS, Hurwitz JL. Vaccine Design Informed by Virus-Induced Immunity. Viral Immunol 2020; 33:342-350. [PMID: 32366204 PMCID: PMC7247049 DOI: 10.1089/vim.2019.0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
When an individual is exposed to a viral pathogen for the first time, the adaptive immune system is naive and cannot prevent virus replication. The consequence may be severe disease. At the same time, the host may rapidly generate a pathogen-specific immune response that will prevent disease if the virus is encountered again. Parvovirus B19 provides one such example. Children with sickle cell disease can experience life-threatening transient aplastic crisis when first exposed to parvovirus B19, but an effective immune response confers lifelong protection. We briefly examine the induction and benefits of virus-induced immunity. We focus on three human viruses for which there are no licensed vaccines (respiratory syncytial virus, human immunodeficiency virus type 1, and parvovirus B19) and consider how virus-induced immunity may inform successful vaccine design.
Collapse
Affiliation(s)
- Rhiannon R. Penkert
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jane S. Hankins
- Pathology Department, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Neal S. Young
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
39
|
Abstract
HIV infection can be effectively treated by lifelong administration of combination antiretroviral therapy, but an effective vaccine will likely be required to end the HIV epidemic. Although the majority of current vaccine strategies focus on the induction of neutralizing antibodies, there is substantial evidence that cellular immunity mediated by CD8+ T cells can sustain long-term disease-free and transmission-free HIV control and may be harnessed to induce both therapeutic and preventive antiviral effects. In this Review, we discuss the increasing evidence derived from individuals who spontaneously control infection without antiretroviral therapy as well as preclinical immunization studies that provide a clear rationale for renewed efforts to develop a CD8+ T cell-based HIV vaccine in conjunction with B cell vaccine efforts. Further, we outline the remaining challenges in translating these findings into viable HIV prevention, treatment and cure strategies. Recently, antibody-mediated control of HIV infection has received considerable attention. Here, the authors discuss the importance of CD8+ T cells in HIV infection and suggest that efforts to develop vaccines that target these cells in conjunction with B cells should be renewed.
Collapse
|
40
|
McBrien JB, Mavigner M, Franchitti L, Smith SA, White E, Tharp GK, Walum H, Busman-Sahay K, Aguilera-Sandoval CR, Thayer WO, Spagnuolo RA, Kovarova M, Wahl A, Cervasi B, Margolis DM, Vanderford TH, Carnathan DG, Paiardini M, Lifson JD, Lee JH, Safrit JT, Bosinger SE, Estes JD, Derdeyn CA, Garcia JV, Kulpa DA, Chahroudi A, Silvestri G. Robust and persistent reactivation of SIV and HIV by N-803 and depletion of CD8 + cells. Nature 2020; 578:154-159. [PMID: 31969705 PMCID: PMC7580846 DOI: 10.1038/s41586-020-1946-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 12/12/2019] [Indexed: 11/09/2022]
Abstract
Human immunodeficiency virus (HIV) persists indefinitely in individuals with HIV who receive antiretroviral therapy (ART) owing to a reservoir of latently infected cells that contain replication-competent virus1-4. Here, to better understand the mechanisms responsible for latency persistence and reversal, we used the interleukin-15 superagonist N-803 in conjunction with the depletion of CD8+ lymphocytes in ART-treated macaques infected with simian immunodeficiency virus (SIV). Although N-803 alone did not reactivate virus production, its administration after the depletion of CD8+ lymphocytes in conjunction with ART treatment induced robust and persistent reactivation of the virus in vivo. We found viraemia of more than 60 copies per ml in all macaques (n = 14; 100%) and in 41 out of a total of 56 samples (73.2%) that were collected each week after N-803 administration. Notably, concordant results were obtained in ART-treated HIV-infected humanized mice. In addition, we observed that co-culture with CD8+ T cells blocked the in vitro latency-reversing effect of N-803 on primary human CD4+ T cells that were latently infected with HIV. These results advance our understanding of the mechanisms responsible for latency reversal and lentivirus reactivation during ART-suppressed infection.
Collapse
Affiliation(s)
- Julia Bergild McBrien
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Lavinia Franchitti
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - S Abigail Smith
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Erick White
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Gregory K Tharp
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Hasse Walum
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Christian R Aguilera-Sandoval
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William O Thayer
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rae Ann Spagnuolo
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Martina Kovarova
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angela Wahl
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Barbara Cervasi
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - David M Margolis
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- University of North Carolina HIV Cure Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas H Vanderford
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Diane G Carnathan
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Mirko Paiardini
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | - Steven E Bosinger
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Cynthia A Derdeyn
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - J Victor Garcia
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Deanna A Kulpa
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Emory + Children's Center for Childhood Infections and Vaccines, Atlanta, GA, USA
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
41
|
Role of Dendritic Cells in Exposing Latent HIV-1 for the Kill. Viruses 2019; 12:v12010037. [PMID: 31905690 PMCID: PMC7019604 DOI: 10.3390/v12010037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/11/2022] Open
Abstract
The development of effective yet nontoxic strategies to target the latent human immunodeficiency virus-1 (HIV-1) reservoir in antiretroviral therapy (ART)-suppressed individuals poses a critical barrier to a functional cure. The ‘kick and kill’ approach to HIV eradication entails proviral reactivation during ART, coupled with generation of cytotoxic T lymphocytes (CTLs) or other immune effectors equipped to eliminate exposed infected cells. Pharmacological latency reversal agents (LRAs) that have produced modest reductions in the latent reservoir ex vivo have not impacted levels of proviral DNA in HIV-infected individuals. An optimal cure strategy incorporates methods that facilitate sufficient antigen exposure on reactivated cells following the induction of proviral gene expression, as well as the elimination of infected targets by either polyfunctional HIV-specific CTLs or other immune-based strategies. Although conventional dendritic cells (DCs) have been used extensively for the purpose of inducing antigen-specific CTL responses in HIV-1 clinical trials, their immunotherapeutic potential as cellular LRAs has been largely ignored. In this review, we discuss the challenges associated with current HIV-1 eradication strategies, as well as the unharnessed potential of ex vivo-programmed DCs for both the ‘kick and kill’ of latent HIV-1.
Collapse
|
42
|
Okoye AA, DeGottardi MQ, Fukazawa Y, Vaidya M, Abana CO, Konfe AL, Fachko DN, Duell DM, Li H, Lum R, Gao L, Park BS, Skalsky RL, Lewis AD, Axthelm MK, Lifson JD, Wong SW, Picker LJ. Role of IL-15 Signaling in the Pathogenesis of Simian Immunodeficiency Virus Infection in Rhesus Macaques. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:2928-2943. [PMID: 31653683 PMCID: PMC6864325 DOI: 10.4049/jimmunol.1900792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/30/2019] [Indexed: 01/04/2023]
Abstract
Although IL-15 has been implicated in the pathogenic hyperimmune activation that drives progressive HIV and SIV infection, as well as in the generation of HIV/SIV target cells, it also supports NK and T cell homeostasis and effector activity, potentially benefiting the host. To understand the role of IL-15 in SIV infection and pathogenesis, we treated two cohorts of SIVmac239-infected rhesus macaques (RM; Macaca mulatta), one with chronic infection, the other with primary infection, with a rhesusized, IL-15-neutralizing mAb (versus an IgG isotype control) for up to 10 wk (n = 7-9 RM per group). In both cohorts, anti-IL-15 was highly efficient at blocking IL-15 signaling in vivo, causing 1) profound depletion of NK cells in blood and tissues throughout the treatment period; 2) substantial, albeit transient, depletion of CD8+ effector memory T cells (TEM) (but not the naive and central memory subsets); and 3) CD4+ and CD8+ TEM hyperproliferation. In primary infection, reduced frequencies of SIV-specific effector T cells in an extralymphoid tissue site were also observed. Despite these effects, the kinetics and extent of SIV replication, CD4+ T cell depletion, and the onset of AIDS were comparable between anti-IL-15- and control-treated groups in both cohorts. However, RM treated with anti-IL-15 during primary infection manifested accelerated reactivation of RM rhadinovirus. Thus, IL-15 support of NK cell and TEM homeostasis does not play a demonstrable, nonredundant role in SIV replication or CD4+ T cell deletion dynamics but may contribute to immune control of oncogenic γ-herpesviruses.
Collapse
Affiliation(s)
- Afam A Okoye
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Maren Q DeGottardi
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Yoshinori Fukazawa
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Mukta Vaidya
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Chike O Abana
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Audrie L Konfe
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Devin N Fachko
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Derick M Duell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - He Li
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Richard Lum
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Lina Gao
- Division of Biostatistics, Department of Public Health and Preventative Medicine, Oregon Health & Science University, Portland, OR 97239; and
| | - Byung S Park
- Division of Biostatistics, Department of Public Health and Preventative Medicine, Oregon Health & Science University, Portland, OR 97239; and
| | - Rebecca L Skalsky
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Anne D Lewis
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Scott W Wong
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006;
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| |
Collapse
|
43
|
Migueles SA, Chairez C, Lin S, Gavil NV, Rosenthal DM, Pooran M, Natarajan V, Rupert A, Dewar R, Rehman T, Sherman BT, Adelsberger J, Leitman SF, Stroncek D, Morse CG, Connors M, Lane HC, Kovacs JA. Adoptive lymphocyte transfer to an HIV-infected progressor from an elite controller. JCI Insight 2019; 4:130664. [PMID: 31415245 PMCID: PMC6795294 DOI: 10.1172/jci.insight.130664] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/12/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDHIV-infected patients with poor virologic control and multidrug-resistant virus have limited therapeutic options. The current study was undertaken to evaluate the safety, immunologic effects, and antiviral activity of peripheral lymphocytes transferred from an elite controller, whose immune system is able to control viral replication without antiretroviral medications, to an HLA-B*2705-matched progressor.METHODSApproximately 22 billion cells were collected from an elite controller by lymphapheresis and infused within 6 hours into a recipient with a preinfusion CD4+ T cell count of 10 cells/μL (1%) and HIV plasma viral load of 114,993 copies/mL.RESULTSDonor cells were cleared from the recipient's peripheral blood by day 8. A transient decrease in viral load to 58,421 (day 3) was followed by a rebound to 702,972 (day 6) before returning to baseline values by day 8. The decreased viral load was temporally associated with peak levels of donor T cells, including CD8+ T cells that had high levels of expression of Ki67, perforin, and granzyme B. Notably, recipient CD8+ T cells also showed increased expression of these markers, especially in HIV-specific tetramer-positive cells.CONCLUSIONThese results suggest that the adoptive transfer of lymphocytes from an HIV-infected elite controller to an HIV-infected patient with progressive disease may be able to perturb the immune system of the recipient in both positive and negative ways.TRIAL REGISTRATIONClinicalTrials.gov NCT00559416.FUNDINGIntramural Research Programs of the US NIH Clinical Center and the National Institute of Allergy and Infectious Diseases (NIAID); the National Cancer Institute.
Collapse
Affiliation(s)
- Stephen A. Migueles
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Cheryl Chairez
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Siying Lin
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Noah V. Gavil
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Danielle M. Rosenthal
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Milad Pooran
- Critical Care Medicine Department, NIH Clinical Center, NIH, Bethesda, Maryland, USA
| | - Ven Natarajan
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Adam Rupert
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Robin Dewar
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Tauseef Rehman
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Brad T. Sherman
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Joseph Adelsberger
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Susan F. Leitman
- Department of Transfusion Medicine, NIH Clinical Center, NIH, Bethesda, Maryland, USA
| | - David Stroncek
- Department of Transfusion Medicine, NIH Clinical Center, NIH, Bethesda, Maryland, USA
| | - Caryn G. Morse
- Critical Care Medicine Department, NIH Clinical Center, NIH, Bethesda, Maryland, USA
| | - Mark Connors
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - H. Clifford Lane
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Joseph A. Kovacs
- Critical Care Medicine Department, NIH Clinical Center, NIH, Bethesda, Maryland, USA
| |
Collapse
|
44
|
Gulla SK, Rao BR, Moku G, Jinka S, Nimmu NV, Khalid S, Patra CR, Chaudhuri A. In vivo targeting of DNA vaccines to dendritic cells using functionalized gold nanoparticles. Biomater Sci 2019; 7:773-788. [PMID: 30601510 DOI: 10.1039/c8bm01272e] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The clinical success of dendritic cell (DC)-based genetic immunization remains critically dependent on the availability of effective and safe nano-carriers for targeting antigen-encoded DNA vaccines to DCs, the most potent antigen-presenting cells in the human body in vivo. Recent studies revealed the efficacies of mannose receptor-mediated in vivo DC-targeted genetic immunization by liposomal DNA vaccine carriers containing both mannose-mimicking shikimoyl and transfection enhancing guanidinyl functionalities. However, to date, the efficacies of this approach have not been examined for metal-based nanoparticle DNA vaccine carriers. Herein, we report for the first time, the design, synthesis, physico-chemical characterization and bioactivities of gold nanoparticles covalently functionalized with a thiol ligand containing both shikimoyl and guanidinyl functionalities (Au-SGSH). We show that Au-SGSH nanoparticles can deliver DNA vaccines to mouse DCs under in vivo conditions. Subcutaneous administration of near infrared (NIR) dye-labeled Au-SGSH showed significant accumulation of the NIR dye in the DCs of the nearby lymph nodes compared to that for the non-targeting NIR-labeled Au-GSH nanoconjugate containing only a covalently tethered guanidinyl group, not the shikimoyl-functionality. Under prophylactic settings, in vivo immunization (s.c.) with the Au-SGSH-pCMV-MART1 nanoplex induced a long-lasting (180 days) immune response against murine melanoma. Notably, mannose receptor-mediated in vivo DC-targeted immunization (s.c.) with the Au-SGSH-MART1 nanoplex significantly inhibited established melanoma growth and increased the overall survivability of melanoma-bearing mice under therapeutic settings. The Au-SGSH nanoparticles reported herein have potential use for in vivo DC-targeted genetic immunization against cancer and infectious diseases.
Collapse
Affiliation(s)
- Suresh Kumar Gulla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Cao Y, Cartwright EK, Silvestri G, Perelson AS. CD8+ lymphocyte control of SIV infection during antiretroviral therapy. PLoS Pathog 2018; 14:e1007350. [PMID: 30308068 PMCID: PMC6199003 DOI: 10.1371/journal.ppat.1007350] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/23/2018] [Accepted: 09/21/2018] [Indexed: 12/19/2022] Open
Abstract
CD8+ lymphocytes play an important role in suppressing in vivo viral replication in HIV infection. However, both the extent to which and the mechanisms by which CD8+ lymphocytes contribute to viral control are not completely understood. A recent experiment depleted CD8+ lymphocytes in simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs) on antiretroviral treatment (ART) to study the role of CD8+ lymphocytes. CD8+ lymphocytes depletion resulted in temporary plasma viremia in all studied RMs. Viral control was restored when CD8+ lymphocytes repopulated. We developed a viral dynamic model to fit the viral load (VL) data from the CD8 depletion experiment. We explicitly modeled the dynamics of the latent reservoir and the SIV-specific effector cell population including their exhaustion and their potential cytolytic and noncytolytic functions. We found that the latent reservoir significantly contributes to the size of the peak VL after CD8 depletion, while drug efficacy plays a lesser role. Our model suggests that the overall CD8+ lymphocyte cytolytic killing rate is dynamically changing depending on the levels of antigen-induced effector cell activation and exhaustion. Based on estimated parameters, our model suggests that before ART or without ART the overall CD8 cytolytic killing rate is small due to exhaustion. However, after the start of ART, the overall CD8 cytolytic killing rate increases due to an expansion of SIV-specific CD8 effector cells. Further, we estimate that the cytolytic killing rate can be significantly larger than the cytopathic death rate in some animals during the second phase of ART-induced viral decay. Lastly, our model provides a new explanation for the puzzling findings by Klatt et al. and Wong et al. that CD8 depletion done immediately before ART has no noticeable effect on the first phase viral decay slope seen after ART initiation Overall, by incorporating effector cells and their exhaustion, our model can explain the effects of CD8 depletion on VL during ART, reveals a detailed dynamic role of CD8+ lymphocytes in controlling viral infection, and provides a unified explanation for CD8 depletion experimental data. CD8+ lymphocytes play an important role in suppressing in vivo viral replication in HIV infection. However, both the extent to which and the mechanisms by which CD8+ lymphocytes contribute to viral control are not completely understood. By mathematically modeling data from a recent CD8 depletion experiment done in antiretroviral (ART) treated animals, our results suggest that the overall CD8+ lymphocyte cytolytic killing rate is dynamically changing depending on the levels of antigen-induced effector cell activation and exhaustion, i.e. before ART or without ART the overall CD8 cytolytic killing rate is small due to exhaustion. However, after the start of ART, the overall CD8 cytolytic killing rate increases due to an expansion of SIV-specific CD8 effector cells. By incorporating effector cells and their exhaustion, our model explains the effects on viral load of CD8 depletion done before ART or during ART, reveals a detailed dynamic role of CD8+ lymphocytes in controlling viral infection, and provides a unified explanation for CD8 depletion experimental data.
Collapse
Affiliation(s)
- Youfang Cao
- Theoretical Biology and Biophysics (T-6), Los Alamos National Laboratory, Los Alamos, NM, United States of America
- Center for Nonlinear Studies (CNLS), Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Emily K. Cartwright
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Alan S. Perelson
- Theoretical Biology and Biophysics (T-6), Los Alamos National Laboratory, Los Alamos, NM, United States of America
- * E-mail:
| |
Collapse
|
46
|
Cardozo EF, Apetrei C, Pandrea I, Ribeiro RM. The dynamics of simian immunodeficiency virus after depletion of CD8+ cells. Immunol Rev 2018; 285:26-37. [PMID: 30129200 PMCID: PMC6352983 DOI: 10.1111/imr.12691] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human immunodeficiency virus infection is still one of the most important causes of morbidity and mortality in the world, with a disproportionate human and economic burden especially in poorer countries. Despite many years of intense research, an aspect that still is not well understood is what (immune) mechanisms control the viral load during the prolonged asymptomatic stage of infection. Because CD8+ T cells have been implicated in this control by multiple lines of evidence, there has been a focus on understanding the potential mechanisms of action of this immune effector population. One type of experiment used to this end has been depleting these cells with monoclonal antibodies in the simian immunodeficiency virus-macaque model and then studying the effect of that depletion on the viral dynamics. Here we review what these experiments have told us. We emphasize modeling studies to interpret the changes in viral load observed in these experiments, including discussion of alternative models, assumptions and interpretations, as well as potential future experiments.
Collapse
Affiliation(s)
- Erwing Fabian Cardozo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
- Laboratorio de Biomatematica, Faculdade de Medicina da Universidade de Lisboa, Portugal
| |
Collapse
|
47
|
Bhattacharyya M, Whitney JB, Seaman M, Barouch DH, Penaloza-MacMaster P. T-cell subset differentiation and antibody responses following antiretroviral therapy during simian immunodeficiency virus infection. Immunology 2018; 155:458-466. [PMID: 30014618 DOI: 10.1111/imm.12985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/29/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022] Open
Abstract
Antiretroviral therapy (ART) for the treatment of human immunodeficiency virus (HIV) infection represents a major breakthrough in the treatment of HIV/acquired immune-deficiency syndrome. However, it remains unclear how ART influences virus-specific immune responses and understanding this is important for developing novel cure and eradication interventions for HIV-1. In the present study, we evaluate how ART impacts T-cell and antibody responses in simian immunodeficiency virus (SIV) -infected rhesus macaques. We evaluated CD4 and CD8 T-cell responses by multiparameter flow cytometry, viral loads by quantitative RT-PCR by a two-step process using SIV-specific primers and antibody neutralization function by luciferase-based TZM-bl assays. We demonstrate that macaques treated with ART exhibit phenotypic and qualitative effects on T-cell and antibody responses. Macaques on ART exhibited low numbers of virus-specific T-cell responses, and these responses appeared to be partially biased towards central memory subsets. More importantly, there were significantly reduced neutralizing antibody responses in macaques treated with ART. Collectively, these data improve the understanding of how virus-specific immune responses are generated during ART, and suggest the potential importance of therapeutic vaccines to maintain adaptive immunity during treated infection.
Collapse
Affiliation(s)
- Mitra Bhattacharyya
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James B Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michael Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
48
|
Abstract
HIV-infected individuals who maintain control of virus without antiretroviral therapy (ART) are called HIV controllers. The immune responses of these individuals suppress HIV viral replication to low levels or, in the case of elite controllers, to undetectable levels. Although some research indicates a role for inferior virulence of the infecting viral strain in natural control, perhaps by way of defective Nef protein function, we find that the majority of research in HIV controllers highlights CD8 T cells as the main suppressor of viral replication. The most convincing evidence for this argument lies in the strong correlation between certain HLA-I alleles, especially B*57, and HIV control status, a finding that has been replicated by many groups. However, natural control can also occur in individuals lacking these specific HLA alleles, and our understanding of what constitutes an effective CD8 T-cell response remains an incomplete picture. Recent research has broadened our understanding of natural HIV control by illustrating the interactions between different immune cells, including innate immune effectors and antigen-presenting cells. For many years, the immune responses of the natural HIV controllers have been studied for clues on how to achieve functional cure in the rest of the HIV-infected population. The goal of a future functional cure to HIV is one where HIV-infected individuals’ immune responses are able to suppress virus long-term without requiring ART. This review highlights recent advances in our understanding of how HIV controllers’ natural immune responses are able to suppress virus.
Collapse
Affiliation(s)
- Sushma Boppana
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Paul Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
| |
Collapse
|
49
|
Dynamics of Simian Immunodeficiency Virus Two-Long-Terminal-Repeat Circles in the Presence and Absence of CD8 + Cells. J Virol 2018; 92:JVI.02100-17. [PMID: 29643246 DOI: 10.1128/jvi.02100-17] [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: 12/02/2017] [Accepted: 04/08/2018] [Indexed: 12/28/2022] Open
Abstract
CD8+ cells play a key role in human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection, but their specific mechanism(s) of action in controlling the virus is unclear. Two-long-terminal-repeat (2-LTR) circles are extrachromosomal products generated upon failed integration of HIV/SIV. To understand the specific effects of CD8+ cells on infected cells, we analyzed the dynamics of 2-LTR circles in SIVmac251-infected rhesus macaques (RMs) treated with an integrase inhibitor (INT). Twenty RMs underwent CD8+ cell depletion and received raltegravir (RAL) monotherapy or a combination of both. Blood, lymph nodes (LNs), and gut biopsy specimens were routinely sampled. Plasma viral loads (pVLs) and 2-LTR circles from peripheral blood mononuclear cells (PBMCs) and LN lymphocytes were measured with quantitative reverse transcription-PCR (qRT-PCR). In the CD8 depletion group, an ∼1-log increase in pVLs and a slow increase in PBMC 2-LTRs occurred following depletion. In the INT group, a strong decline in pVLs upon treatment initiation and no change in 2-LTR levels were observed. In the INT and CD8+ cell depletion group, an increase in pVLs following CD8 depletion similar to that in the CD8 depletion group was observed, with a modest decline following INT initiation, and 2-LTR circles significantly increased in PBMCs and LNs. Analyzing the 2-LTR data across all treatment groups with a mathematical model indicates that the data best support an effect of CD8+ cells in killing cells prior to viral integration. Sensitivity analyses of these results confirm that effect but also allow for additional effects, which the data do not discriminate well. Overall, we show that INT does not significantly increase the levels of 2-LTR circles. However, CD8+ cell depletion increases the 2-LTR levels, which are enhanced in the presence of an INT.IMPORTANCE CD8+ T cells play an essential role in controlling HIV and SIV infection, but the specific mechanisms involved remain poorly understood. Due to failed viral infection, HIV and SIV can form 2-LTR extrachromosomal circles that can be quantified. We present novel data on the dynamics of these 2-LTR forms in a SIV-infected macaque model under three different treatment conditions: depletion of CD8+ cells, administration of the integrase inhibitor in a monotherapy, which favors the formation of 2-LTR circles, and a combination of the two treatments. We used a new mathematical model to help interpret the data, and the results suggest that CD8+ cells exert a killing effect on infected cells prior to virus integration. These results provide new insights into the mechanisms of action of CD8+ cells in SIV infection. Confirmation of our results would be an important step in understanding immune control of HIV.
Collapse
|
50
|
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
|