1
|
Chéret A. Acute HIV-1 Infection: Paradigm and Singularity. Viruses 2025; 17:366. [PMID: 40143294 PMCID: PMC11945883 DOI: 10.3390/v17030366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Acute HIV-1 infection (AHI) is a transient period where the virus causes evident damage to the immune system, including an extensive apoptosis of CD4+ T cells associated with a high level of activation and a major cytokine storm to fight the invading virus. HIV infection establishes persistence by integrating the viral genome into host cell DNA in both replicating and non-replicating forms, effectively hiding from immune surveillance within infected lymphocytes as cellular reservoirs. The measurement of total HIV-1 DNA in peripheral blood mononuclear cells (PBMCs) is a reliable reflection of this reservoir. Initiating treatments during AHI with nucleoside reverse transcriptase inhibitors (NRTIs) and/or integrase strand transfer inhibitors (INSTIs) is essential to alter the dynamics of the global reservoir expansion, and to reduce the establishment of long-lived cellular and tissue reservoirs, while preserving and enhancing specific and non-specific immune responses. Furthermore, some of the patients treated at the AHI stage may become post-treatment controllers and should be informative regarding the mechanism of viral control, so patients treated during AHI are undoubtedly the best candidates to test innovative remission strategies toward a functional cure that could play a pivotal role in long-term HIV control. AHI is characterized by high levels of viral replication, with a significant increase in the risk of HIV transmission. Detecting AHI and initiating early treatment following diagnosis provides a window of opportunity to control the epidemic, particularly in high-risk populations.
Collapse
Affiliation(s)
- Antoine Chéret
- Inserm U1016, CNRS UMR 8104, Institut Cochin, Université Paris Descartes, 75014 Paris, France;
- Service Plateforme de Diagnostic et Thérapeutique Pluridisciplinaire, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
- INSERM-CIC-1424, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
| |
Collapse
|
2
|
Li L, Zhao M, van Meurs M, Brouwers-Haspels I, den Dekker RJH, Wilmsen MEP, Grashof DGB, van de Werken HJG, Rao S, Rokx C, Mueller YM, Katsikis PD. Bryostatin-1 enhances the proliferation and functionality of exhausted CD8+ T cells by upregulating MAP Kinase 11. Front Immunol 2025; 15:1509874. [PMID: 39877358 PMCID: PMC11772198 DOI: 10.3389/fimmu.2024.1509874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Bryostatin-1, a potent agonist of the protein kinase C, has been studied for HIV and cancer therapies. In HIV research, it has shown anti-HIV effects during acute infection and reactivation of latent HIV in chronic infection. As effective CD8+ T cell responses are essential for eliminating reactivated virus and achieving a cure, it is important to investigate how bryostatin-1 affects HIV-specific CD8+ T cells. HIV-specific CD8+ T cells often become exhausted, showing reduced proliferative potential and impaired cytokine production, a dysfunction also observed in cancer. Therefore, we further investigated how bryostatin-1 directly impacts exhausted CD8+ T cells. Methods PBMCs from people with HIV (PWH) were treated with bryostatin-1 and tracked with proliferation dye for cell expansion. One day 6, HIV-specific CD8+ T cells were detected by tetramers staining and examined by flow cytometry. By utilizing an established in vitro murine T cell exhaustion system, changes in inhibitory receptors, transcription factors, cytokine production and killing capacity of bryostatin-1 treated exhausted CD8+ T cells were determined by flow cytometry. RNA-seq analysis was performed to study transcriptional changes in these cells. Results We found that bryostatin-1 improved the expansion and decreased PD-1 expression of HIV-specific CD8+ T cells. Bryostatin-1 enhanced the functionality and proliferation while decreasing inhibitory receptor expression of in vitro generated exhausted CD8+ T cells. Bryostatin-1 upregulated TCF-1 and decreased TOX expression. These changes were confirmed through RNA-seq analysis. RNA-seq revealed that mitogen-activated protein kinases (MAPK) 11 was significantly downregulated in exhausted CD8+ T cells, however, it greatly upregulated after bryostatin-1 treatment. Inhibition of MAPK11 in bryostatin-1-treated cells blocked the increased proliferation and IFN-γ production induced by bryostatin-1, but did not affect other bryostatin-1 induced effects, such as the reduction of inhibitory receptors. Discussion Our data demonstrate that bryostatin-1 induces a MAPK 11-dependent improvement in the proliferative and functional capacity of exhausted T cells. This study provides a rationale for bryostatin-1's potential to help eradicate the HIV reservoir during treatment, and it may also contribute to cancer immunotherapy by functionally improving exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Ling Li
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Manzhi Zhao
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marjan van Meurs
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | | | - Merel E. P. Wilmsen
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dwin G. B. Grashof
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Shringar Rao
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Casper Rokx
- Department of Internal Medicine, and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yvonne M. Mueller
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Peter D. Katsikis
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
3
|
Abstract
Antiretroviral therapies efficiently block HIV-1 replication but need to be maintained for life. Moreover, chronic inflammation is a hallmark of HIV-1 infection that persists despite treatment. There is, therefore, an urgent need to better understand the mechanisms driving HIV-1 pathogenesis and to identify new targets for therapeutic intervention. In the past few years, the decisive role of cellular metabolism in the fate and activity of immune cells has been uncovered, as well as its impact on the outcome of infectious diseases. Emerging evidence suggests that immunometabolism has a key role in HIV-1 pathogenesis. The metabolic pathways of CD4+ T cells and macrophages determine their susceptibility to infection, the persistence of infected cells and the establishment of latency. Immunometabolism also shapes immune responses against HIV-1, and cell metabolic products are key drivers of inflammation during infection. In this Review, we summarize current knowledge of the links between HIV-1 infection and immunometabolism, and we discuss the potential opportunities and challenges for therapeutic interventions.
Collapse
|
4
|
Ndhlovu ZM, Kazer SW, Nkosi T, Ogunshola F, Muema DM, Anmole G, Swann SA, Moodley A, Dong K, Reddy T, Brockman MA, Shalek AK, Ndung'u T, Walker BD. Augmentation of HIV-specific T cell function by immediate treatment of hyperacute HIV-1 infection. Sci Transl Med 2020; 11:11/493/eaau0528. [PMID: 31118290 DOI: 10.1126/scitranslmed.aau0528] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/31/2018] [Accepted: 03/28/2019] [Indexed: 12/13/2022]
Abstract
Sustained viremia after acute HIV infection is associated with profound CD4+ T cell loss and exhaustion of HIV-specific CD8+ T cell responses. To determine the impact of combination antiretroviral therapy (cART) on these processes, we examined the evolution of immune responses in acutely infected individuals initiating treatment before peak viremia. Immediate treatment of Fiebig stages I and II infection led to a rapid decline in viral load and diminished magnitude of HIV-specific (tetramer+) CD8+ T cell responses compared to untreated donors. There was a strong positive correlation between cumulative viral antigen exposure before full cART-induced suppression and immune responses measured by MHC class I tetramers, IFN-γ ELISPOT, and CD8+ T cell activation. HIV-specific CD8+ T responses of early treated individuals were characterized by increased CD127 and BCL-2 expression, greater in vitro IFN-γ secretion, and enhanced differentiation into effector memory (Tem) cells. Transcriptional analysis of tetramer+ CD8+ T cells from treated persons revealed reduced expression of genes associated with activation and apoptosis, with concurrent up-regulation of prosurvival genes including BCL-2, AXL, and SRC Early treatment also resulted in robust HIV-specific CD4+ T cell responses compared to untreated HIV-infected individuals. Our data show that limiting acute viremia results in enhanced functionality of HIV-specific CD4+ and CD8+ T cells, preserving key antiviral properties of these cells.
Collapse
Affiliation(s)
- Zaza M Ndhlovu
- Africa Health Research Institute, 4036 Durban, South Africa. .,Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 4013 Durban, South Africa
| | - Samuel W Kazer
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA 02139, USA.,Department of Chemistry and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Thandeka Nkosi
- Africa Health Research Institute, 4036 Durban, South Africa.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 4013 Durban, South Africa
| | - Funsho Ogunshola
- Africa Health Research Institute, 4036 Durban, South Africa.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 4013 Durban, South Africa
| | - Daniel M Muema
- Africa Health Research Institute, 4036 Durban, South Africa
| | - Gursev Anmole
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Shayda A Swann
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Amber Moodley
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 4013 Durban, South Africa
| | - Krista Dong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Tarylee Reddy
- South Africa Medical Research Council, 4091 Durban, South Africa
| | - Mark A Brockman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA 02139, USA.,Department of Chemistry and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Thumbi Ndung'u
- Africa Health Research Institute, 4036 Durban, South Africa.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 4013 Durban, South Africa
| | - Bruce D Walker
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 4013 Durban, South Africa.,Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
5
|
Song CB, Zhang LL, Wu X, Fu YJ, Jiang YJ, Shang H, Zhang ZN. CD4 +CD38 + central memory T cells contribute to HIV persistence in HIV-infected individuals on long-term ART. J Transl Med 2020; 18:95. [PMID: 32093678 PMCID: PMC7038621 DOI: 10.1186/s12967-020-02245-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background Despite the effective antiretroviral treatment (ART) of HIV-infected individuals, HIV persists in a small pool. Central memory CD4+ T cells (Tcm) make a major contribution to HIV persistence. We found that unlike HLA-DR, CD38 is highly expressed on the Tcm of HIV-infected subjects receiving ART for > 5 years. It has been reported that the half-life of total and episomal HIV DNA in the CD4+CD38+ T cell subset, exhibits lower decay rates at 12 weeks of ART. Whether CD38 contributes to HIV latency in HIV-infected individuals receiving long-term ART is yet to be addressed. Methods Peripheral blood mononuclear cells (PBMCs) were isolated from the whole blood of HIV-infected subjects receiving suppressive ART. The immunophenotyping, proliferation and apoptosis of CD4+ T cell subpopulations were detected by flow cytometry, and the level of CD38 mRNA and total HIV DNA were measured using real-time PCR and digital droplet PCR, respectively. A negative binomial regression model was used to determine the correlation between CD4+CD38+ Tcm and total HIV DNA in CD4+ T cells. Results CD38 was highly expressed on CD4+ Tcm cells from HIV infected individuals on long-term ART. Comparing with HLA-DR−Tcm and CD4+HLA-DR+ T cells, CD4+CD38+ Tcm cells displayed lower levels of activation (CD25 and CD69) and higher levels of CD127 expression. The proportion of CD38+ Tcm, but not CD38− Tcm cells can predict the total HIV DNA in the CD4+ T cells and the CD38+ Tcm subset harbored higher total HIV DNA copy numbers than the CD38− Tcm subset. After transfected with CD38 si-RNA in CD4+ T cells, the proliferation of CD4+ T cells was inhibited. Conclusion The current date indicates that CD4+CD38+ Tcm cells contribute to HIV persistence in HIV-infected individuals on long-term ART. Our study provides a potential target to resolve HIV persistence.
Collapse
Affiliation(s)
- Cheng-Bo Song
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Le-Le Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Xian Wu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Ya-Jing Fu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Yong-Jun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China. .,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China.
| | - Zi-Ning Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjingbei Street, Heping District, Shenyang, 110001, Liaoning Province, China. .,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China.
| |
Collapse
|
6
|
Chandrasekar AP, Cummins NW, Badley AD. The Role of the BCL-2 Family of Proteins in HIV-1 Pathogenesis and Persistence. Clin Microbiol Rev 2019; 33:e00107-19. [PMID: 31666279 PMCID: PMC6822993 DOI: 10.1128/cmr.00107-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Advances in HIV-1 therapy have transformed the once fatal infection into a manageable, chronic condition, yet the search for a widely applicable approach to cure remains elusive. The ineffectiveness of antiretroviral therapy (ART) in reducing the size of the HIV-1 latent reservoir has prompted investigation into the mechanisms of HIV-1 latency and immune escape. One of the major regulators of apoptosis, the BCL-2 protein, alongside its homologous family members, is a major target of HIV-1-induced change. Recent studies have now demonstrated the association of this protein with cells that support proviral forms in the setting of latency and have helped identify BCL-2 as a novel and promising therapeutic target for HIV-1 therapy directed at possible cure. This review aims to systematically review the interactions of HIV-1 with BCL-2 and its homologs and to examine the possibility of using BCL-2 inhibitors in the study and elimination of the latent reservoir.
Collapse
Affiliation(s)
| | - Nathan W Cummins
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Lin TC, Gianella S, Tenenbaum T, Little SJ, Hoenigl M. A Simple Symptom Score for Acute Human Immunodeficiency Virus Infection in a San Diego Community-Based Screening Program. Clin Infect Dis 2019; 67:105-111. [PMID: 29293891 DOI: 10.1093/cid/cix1130] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/23/2017] [Indexed: 12/15/2022] Open
Abstract
Background Treatment of acute human immunodeficiency virus (HIV) infection (AHI) decreases transmission and preserves immune function, but AHI diagnosis remains resource intensive. Risk-based scores predictive for AHI have been described for high-risk groups; however, symptom-based scores could be more generalizable across populations. Methods Adults who tested either positive for AHI (antibody-negative, HIV nucleic acid test [NAT] positive) or HIV NAT negative with the community-based San Diego Early Test HIV screening program were retrospectively randomized 2:1 into a derivation and validation set. In the former, symptoms significant for AHI in a multivariate logistic regression model were assigned a score value (the odds ratio [OR] rounded to the nearest integer). The score was assessed in the validation set using receiver operating characteristics and areas under the curve (AUC). An optimal cutoff score was found using the Youden index. Results Of 998 participants (including 261 non-men who have sex with men [MSM]), 113 had AHI (including 4 non-MSM). Compared to HIV-negative cases, AHI cases reported more symptoms (median, 4 vs 0; P < .01). Fever, myalgia, and weight loss were significantly associated with AHI in the multivariate model and corresponded to 11, 8, and 4 score points, respectively. The summed score yielded an AUC of 0.85 (95% confidence interval [CI], .77-.93). A score of ≥11 was 72% sensitive and 96% specific (diagnostic OR, 70.27). Conclusions A 3-symptom score accurately predicted AHI in a community-based screening program and may inform allocation of resources in settings that do not routinely screen for AHI.
Collapse
Affiliation(s)
- Timothy C Lin
- University of California, San Diego School of Medicine, La Jolla
| | - Sara Gianella
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego
| | - Tara Tenenbaum
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego
| | - Susan J Little
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego
| | - Martin Hoenigl
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego.,Section of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Austria.,Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Austria
| |
Collapse
|
8
|
Hsu DC, Ananworanich J. Immune Interventions to Eliminate the HIV Reservoir. Curr Top Microbiol Immunol 2017; 417:181-210. [PMID: 29071472 DOI: 10.1007/82_2017_70] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inducing HIV remission is a monumental challenge. A potential strategy is the "kick and kill" approach where latently infected cells are first activated to express viral proteins and then eliminated through cytopathic effects of HIV or immune-mediated killing. However, pre-existing immune responses to HIV cannot eradicate HIV infection due to the presence of escape variants, inadequate magnitude, and breadth of responses as well as immune exhaustion. The two major approaches to boost immune-mediated elimination of infected cells include enhancing cytotoxic T lymphocyte mediated killing and harnessing antibodies to eliminate HIV. Specific strategies include increasing the magnitude and breadth of T cell responses through therapeutic vaccinations, reversing the effects of T cell exhaustion using immune checkpoint inhibition, employing bispecific T cell targeting immunomodulatory proteins or dual-affinity re-targeting molecules to direct cytotoxic T lymphocytes to virus-expressing cells and broadly neutralizing antibody infusions. Methods to steer immune responses to tissue sites where latently infected cells are located need to be further explored. Ultimately, strategies to induce HIV remission must be tolerable, safe, and scalable in order to make a global impact.
Collapse
Affiliation(s)
- Denise C Hsu
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Jintanat Ananworanich
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA. .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA. .,US Military HIV Research Program (MHRP), 6720-A Rockledge Drive, Suite 400, Bethesda, MD, 20817, USA.
| |
Collapse
|
9
|
Eberhard JM, Ahmad F, Hong HS, Bhatnagar N, Keudel P, Schulze Zur Wiesch J, Schmidt RE, Meyer-Olson D. Partial recovery of senescence and differentiation disturbances in CD8 + T cell effector-memory cells in HIV-1 infection after initiation of anti-retroviral treatment. Clin Exp Immunol 2016; 186:227-238. [PMID: 27377704 DOI: 10.1111/cei.12837] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/13/2016] [Accepted: 06/24/2016] [Indexed: 01/22/2023] Open
Abstract
Immune senescence as well as disturbed CD8+ T cell differentiation are a hallmark of chronic HIV infection. Here, we investigated to what extent immune senescence is reversible after initiation of anti-retroviral treatment (ART). Peripheral blood mononuclear cells (PBMCs) from a cohort of HIV patients with different disease courses, including untreated viral controllers (n = 10), viral non-controllers (n = 16) and patients on ART (n = 20), were analysed and compared to uninfected controls (n = 25) by flow cytometry on bulk and HIV-specific major histocompatibility complex (MHC) class I tetramer+ CD8+ T cells for expression of the memory markers CCR7 and CD45RO, as well as the senescence marker CD57 and the differentiation and survival marker CD127. Furthermore, a subset of patients was analysed longitudinally before and after initiation of ART. Frequencies of CD57+ CD8+ T cells decreased after initiation of ART in central memory (Tcm) but not in effector memory T cell populations (TemRO and TemRA). The frequency of CD127+ CD8+ cells increased in Tcm and TemRO. We observed a reduction of CD127- T cells in Tcm, TemRO and partially in TemRA subsets after initiation of ART. Importantly, HIV-specific CD8+ TemRO cells predominantly displayed a CD127- CD57+ phenotype in untreated HIV-patients, whereas the CD127+ CD57- phenotype was under-represented in these patients. The frequency of the CD127+ CD57- CD8+ T cell subpopulation correlated strongly with absolute CD4+ counts in HIV-infected patients before and after initiation of ART. These findings can be interpreted as a phenotypical correlate of CD8+ memory T cell differentiation and the premature 'ageing' of the immune system, which was even observed in successfully virally suppressed HIV patients.
Collapse
Affiliation(s)
- J M Eberhard
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany. .,Zentrum für Innere Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| | - F Ahmad
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - H S Hong
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany.,Division of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, MA, USA
| | - N Bhatnagar
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany.,Unit of Cytokines and Inflammation, Institute Pasteur, Paris, France
| | - P Keudel
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - J Schulze Zur Wiesch
- Zentrum für Innere Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.,DZIF German Center for Infection-Partner Sites Hamburg, Hannover, Germany
| | - R E Schmidt
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany.,DZIF German Center for Infection-Partner Sites Hamburg, Hannover, Germany
| | - D Meyer-Olson
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Hannover, Germany
| |
Collapse
|
10
|
Amu S, Lantto Graham R, Bekele Y, Nasi A, Bengtsson C, Rethi B, Sorial S, Meini G, Zazzi M, Hejdeman B, Chiodi F. Dysfunctional phenotypes of CD4+ and CD8+ T cells are comparable in patients initiating ART during early or chronic HIV-1 infection. Medicine (Baltimore) 2016; 95:e3738. [PMID: 27281071 PMCID: PMC4907649 DOI: 10.1097/md.0000000000003738] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Early initiation of antiretroviral therapy (ART) is becoming a common clinical practice according to current guidelines recommending treatment to all HIV-1-infected patients. However, it is not known whether ART initiated during the early phase of infection prevents the establishment of abnormal phenotypic features previously reported in CD4+ and CD8+T cells during chronic HIV-1 infection. In this cross-sectional study, blood specimens were obtained from 17 HIV-1-infected patients who began ART treatment shortly after infection (early ART [EA]), 17 age-matched HIV-1-infected patients who started ART during chronic phase of infection (late ART [LA]), and 25 age-matched non-HIV-1-infected controls. At collection of specimens, patients in EA and LA groups had received ART for comparable periods of time. Total HIV-1 DNA was measured in white blood cells by quantitative PCR. The concentration of 9 inflammatory parameters and 1 marker of fibrosis, including sCD14 and β-2 microglobulin, was measured in plasma. Furthermore, expression of markers of abnormal immune activation (human leukocyte antigen - antigen D related [HLA-DR] and CD38), exhaustion (programmed death 1, CD28, CD57) and terminal differentiation (CD127) was measured on CD4+ and CD8+T cells. T-cell proliferation was measured through Ki67 expression. The copies of total HIV-1 DNA in blood were significantly lower (P = 0.009) in EA compared with that in LA group. Only the expression of HLA-DR on naïve CD4+ T cells distinguished EA from LA, whereas expression of 3 surface markers distinguished T-cell populations of HIV-1-infected patients from controls. These included HLA-DR distinguishing CD4+ T cells from EA compared with controls, and also CD38 and CD127 on CD4+ and CD8+ T cells, respectively, distinguishing both groups of patients from controls. The sCD14 levels were significantly higher in EA patients, and β-2 microglobulin levels were higher in LA group compared with that in controls. Our results demonstrate an equivalent abnormal expression of activation (HLA-DR and CD38 on CD4+ T cells) and terminal differentiation (CD127 on CD8+ T cells) markers in T cells from both EA and LA patients. The size of total HIV-1 DNA copies in blood of EA was lower compared with LA patients. These findings suggest that some abnormalities taking place in the T-cell compartment during primary HIV-1 infection may not be corrected by early ART.
Collapse
Affiliation(s)
- Sylvie Amu
- Department of Microbiology, Tumor and Cell Biology
| | | | - Yonas Bekele
- Department of Microbiology, Tumor and Cell Biology
| | | | | | - Bence Rethi
- Department of Microbiology, Tumor and Cell Biology
- Department of Medicine at Solna
| | - Sam Sorial
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet and Unit of Infectious Diseases/Venhälsan, Södersjukhuset, Stockholm, Sweden
| | - Genny Meini
- Department of Microbiology and Virology, Policlinico S. Maria alle Scotte, Siena, Italy
| | - Maurizio Zazzi
- Department of Microbiology and Virology, Policlinico S. Maria alle Scotte, Siena, Italy
| | - Bo Hejdeman
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet and Unit of Infectious Diseases/Venhälsan, Södersjukhuset, Stockholm, Sweden
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology
- ∗Correspondence: Francesca Chiodi, Department of Microbiology, Tumor and Cell Biology, Nobels väg 16, 17177 Stockholm, Sweden (e-mail: )
| |
Collapse
|
11
|
Holst PJ, Jensen BAH, Ragonnaud E, Thomsen AR, Christensen JP. Targeting of non-dominant antigens as a vaccine strategy to broaden T-cell responses during chronic viral infection. PLoS One 2015; 10:e0117242. [PMID: 25679375 PMCID: PMC4334508 DOI: 10.1371/journal.pone.0117242] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022] Open
Abstract
In this study, we compared adenoviral vaccine vectors with the capacity to induce equally potent immune responses against non-dominant and immunodominant epitopes of murine lymphocytic choriomeningitis virus (LCMV). Our results demonstrate that vaccination targeting non-dominant epitopes facilitates potent virus-induced T-cell responses against immunodominant epitopes during subsequent challenge with highly invasive virus. In contrast, when an immunodominant epitope was included in the vaccine, the T-cell response associated with viral challenge remained focussed on that epitope. Early after challenge with live virus, the CD8+ T cells specific for vaccine-encoded epitopes, displayed a phenotype typically associated with prolonged/persistent antigenic stimulation marked by high levels of KLRG-1, as compared to T cells reacting to epitopes not included in the vaccine. Notably, this association was lost over time in T cells specific for the dominant T cell epitopes, and these cells were fully capable of expanding in response to a new viral challenge. Overall, our data suggests a potential for broadening of the antiviral CD8+ T-cell response by selecting non-dominant antigens to be targeted by vaccination. In addition, our findings suggest that prior adenoviral vaccination is not likely to negatively impact the long-term and protective immune response induced and maintained by a vaccine-attenuated chronic viral infection.
Collapse
Affiliation(s)
- Peter J. Holst
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Benjamin A. H. Jensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Emeline Ragonnaud
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan R. Thomsen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| | - Jan P. Christensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, Copenhagen, Denmark
| |
Collapse
|
12
|
Ambrosioni J, Nicolas D, Sued O, Agüero F, Manzardo C, Miro JM. Update on antiretroviral treatment during primary HIV infection. Expert Rev Anti Infect Ther 2014; 12:793-807. [PMID: 24803105 DOI: 10.1586/14787210.2014.913981] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary HIV-1 infection covers a period of around 12 weeks in which the virus disseminates from the initial site of infection into different tissues and organs. In this phase, viremia is very high and transmission of HIV is an important issue. Most guidelines recommend antiretroviral treatment in patients who are symptomatic, although the indication for treatment remains inconclusive in asymptomatic patients. In this article the authors review the main virological and immunological events during this early phase of infection, and discuss the arguments for and against antiretroviral treatment. Recommendations of different guidelines, the issue of the HIV transmission and transmission of resistance to antiretroviral drugs, as well as recently available information opening perspectives for functional cure in patients treated in very early steps of HIV infection are also discussed.
Collapse
Affiliation(s)
- Juan Ambrosioni
- Infectious Diseases Service, Hospital Clinic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
13
|
Kawana-Tachikawa A, Llibre JM, Bravo I, Escrig R, Mothe B, Puig J, Puertas MC, Martinez-Picado J, Blanco J, Manzardo C, Miro JM, Iwamoto A, Pozniak AL, Gatell JM, Clotet B, Brander C, the MARAVIBOOST investigators. Effect of maraviroc intensification on HIV-1-specific T cell immunity in recently HIV-1-infected individuals. PLoS One 2014; 9:e87334. [PMID: 24475275 PMCID: PMC3903883 DOI: 10.1371/journal.pone.0087334] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/19/2013] [Indexed: 01/22/2023] Open
Abstract
Background The effect of maraviroc on the maintenance and the function of HIV-1-specific T cell responses remains unknown. Methods Subjects recently infected with HIV-1 were randomized to receive anti-retroviral treatment with or without maraviroc intensification for 48 weeks, and were monitored up to week 60. PBMC and in vitro-expanded T cells were tested for responses to the entire HIV proteome by ELISpot analyses. Intracellular cytokine staining assays were conducted to monitor the (poly)-functionality of HIV-1-specific T cells. Analyses were performed at baseline and week 24 after treatment start, and at week 60 (3 months after maraviroc discontinuation). Results Maraviroc intensification was associated with a slower decay of virus-specific T cell responses over time compared to the non-intensified regimen in both direct ex-vivo as well as in in-vitro expanded cells. The effector function profiles of virus-specific CD8+ T cells were indistinguishable between the two arms and did not change over time between the groups. Conclusions Maraviroc did not negatively impact any of the measured parameters, but was rather associated with a prolonged maintenance of HIV-1-specific T cell responses. Maraviroc, in addition to its original effect as viral entry inhibitor, may provide an additional benefit on the maintenance of virus-specific T cells which may be especially important for future viral eradication strategies.
Collapse
Affiliation(s)
- Ai Kawana-Tachikawa
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
- Division of Infectious Diseases, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Josep M. Llibre
- Lluita contra la SIDA Foundation, HIV Unit, University Hospital Germans Trias i Pujol, Badalona, UAB, Badalona, Spain
| | - Isabel Bravo
- Lluita contra la SIDA Foundation, HIV Unit, University Hospital Germans Trias i Pujol, Badalona, UAB, Badalona, Spain
| | - Roser Escrig
- Lluita contra la SIDA Foundation, HIV Unit, University Hospital Germans Trias i Pujol, Badalona, UAB, Badalona, Spain
| | - Beatriz Mothe
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
- Lluita contra la SIDA Foundation, HIV Unit, University Hospital Germans Trias i Pujol, Badalona, UAB, Badalona, Spain
- University of Vic, Vic, Spain
| | - Jordi Puig
- Lluita contra la SIDA Foundation, HIV Unit, University Hospital Germans Trias i Pujol, Badalona, UAB, Badalona, Spain
| | - Maria C. Puertas
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
| | - Javier Martinez-Picado
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
- University of Vic, Vic, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - Julia Blanco
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
- University of Vic, Vic, Spain
| | | | - Jose M. Miro
- Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Aikichi Iwamoto
- Division of Infectious Diseases, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Anton L. Pozniak
- HIV/GUM Department, Chelsea and Westminster Hospital, London, United Kingdom
| | - Jose M. Gatell
- Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
- Lluita contra la SIDA Foundation, HIV Unit, University Hospital Germans Trias i Pujol, Badalona, UAB, Badalona, Spain
- University of Vic, Vic, Spain
| | - Christian Brander
- Irsicaixa AIDS Research Institute – HIVACAT, Autonomous University of Barcelona, Badalona, Spain
- University of Vic, Vic, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
- * E-mail:
| | | |
Collapse
|
14
|
HIV immune activation drives increased Eomes expression in memory CD8 T cells in association with transcriptional downregulation of CD127. AIDS 2013; 27:1867-77. [PMID: 23965471 DOI: 10.1097/qad.0b013e3283618487] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND During HIV infection distinct mechanisms drive immune activation of the CD4 and CD8 T cells leading to CD4 T-cell depletion and expansion of the CD8 T-cell pool. This immune activation is polyclonal and extends beyond HIV-specific T cells. One consequence of this immune activation is a profound decrease in IL-7Rα (CD127) expression on memory CD8 T cells. The mechanisms leading to this are unknown and because of the potential impact of reduced IL-7 signaling in memory T cells specific to HIV and other pathogens, in the present study we examined the molecular mechanisms implicated in this downregulation of CD127. METHODS Membrane bound (mIL7RA) and soluble (sIL7RA) mRNA expression was determined by qRT-PCR. CD127, Eomesodermin (Eomes) and T-bet expression in healthy controls and HIV-infected patients were studied by flow cytometry. RESULTS CD127 downregulation occurs at the transcriptional level for both mIL7RA and sIL7RA alternative spliced forms in the CD127 memory CD8 T cells. CD127 memory CD8 T cells exhibited increased Eomes expression and an 'effector-like' gene profile. These changes were associated with higher HIV-RNA levels. Following combination antiretroviral therapy (cART), there was an increase in CD127 expression over an extended period of time (>5 months) which was associated with decreased Eomes expression. CONCLUSION CD127 is downregulated at a transcriptional level in memory CD8 T cells in association with upregulation of Eomes expression.
Collapse
|
15
|
Sundaravaradan V, Mir KD, Sodora DL. Double-negative T cells during HIV/SIV infections: potential pinch hitters in the T-cell lineup. Curr Opin HIV AIDS 2012; 7:164-71. [PMID: 22241163 PMCID: PMC3639317 DOI: 10.1097/coh.0b013e3283504a66] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW This review summarizes the role of CD3+CD4-CD8- double-negative T cells, which have both regulatory and helper T-cell functions and may have the potential to compensate for the reduced levels of CD4 T cells during SIV/HIV infection. RECENT FINDINGS Double-negative T cells have been characterized in several human diseases and in murine models of autoimmunity and transplantation, where they exhibit both immunoregulatory and helper T-cell-like function. During the natural nonpathogenic SIV infection of African nonhuman primates, the lack of clinical disease progression is associated with the presence of double-negative T cells that maintain helper T-cell functions while remaining refractory to viral infection. Moreover, DN T cells may compensate for very low levels of CD4+ T cells observed in a cohort of SIV-infected sooty mangabeys that have remained free of clinical AIDS for over 10 years. These studies identify a potential for double-negative T cells to provide critical helper function during HIV infection. SUMMARY Double-negative T cells with some CD4+ T-cell functions are associated with a nonpathogenic outcome during SIV infection and represent a potential immune therapeutic target in HIV-infected patients.
Collapse
|
16
|
Goujard C, Girault I, Rouzioux C, Lécuroux C, Deveau C, Chaix ML, Jacomet C, Talamali A, Delfraissy JF, Venet A, Meyer L, Sinet M. HIV-1 control after transient antiretroviral treatment initiated in primary infection: role of patient characteristics and effect of therapy. Antivir Ther 2012; 17:1001-9. [DOI: 10.3851/imp2273] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2012] [Indexed: 10/28/2022]
|
17
|
Cellerai C, Perreau M, Rozot V, Enders FB, Pantaleo G, Harari A. Proliferation capacity and cytotoxic activity are mediated by functionally and phenotypically distinct virus-specific CD8 T cells defined by interleukin-7R{alpha} (CD127) and perforin expression. J Virol 2010; 84:3868-78. [PMID: 20130059 PMCID: PMC2849500 DOI: 10.1128/jvi.02565-09] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 01/21/2010] [Indexed: 11/20/2022] Open
Abstract
Cytotoxicity and proliferation capacity are key functions of antiviral CD8 T cells. In the present study, we investigated a series of markers to define these functions in virus-specific CD8 T cells. We provide evidence that there is a lack of coexpression of perforin and CD127 in human CD8 T cells. CD127 expression on virus-specific CD8 T cells correlated positively with proliferation capacity and negatively with perforin expression and cytotoxicity. Influenza virus-, cytomegalovirus-, and Epstein-Barr virus/human immunodeficiency virus type 1-specific CD8 T cells were predominantly composed of CD127(+) perforin(-)/CD127(-) perforin(+), and CD127(-)/perforin(-) CD8 T cells, respectively. CD127(-)/perforin(-) and CD127(-)/perforin(+) cells expressed significantly more PD-1 and CD57, respectively. Consistently, intracellular cytokine (gamma interferon, tumor necrosis factor alpha, and interleukin-2 [IL-2]) responses combined to perforin detection confirmed that virus-specific CD8 T cells were mostly composed of either perforin(+)/IL-2(-) or perforin(-)/IL-2(+) cells. In addition, perforin expression and IL-2 secretion were negatively correlated in virus-specific CD8 T cells (P < 0.01). As previously shown for perforin, changes in antigen exposure modulated also CD127 expression. Based on the above results, proliferating (CD127(+)/IL-2-secreting) and cytotoxic (perforin(+)) CD8 T cells were contained within phenotypically distinct T-cell populations at different stages of activation or differentiation and showed different levels of exhaustion and senescence. Furthermore, the composition of proliferating and cytotoxic CD8 T cells for a given antiviral CD8 T-cell population appeared to be influenced by antigen exposure. These results advance our understanding of the relationship between cytotoxicity, proliferation capacity, the levels of senescence and exhaustion, and antigen exposure of antiviral memory CD8 T cells.
Collapse
Affiliation(s)
- Cristina Cellerai
- Laboratory of AIDS Immunopathogenesis, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Matthieu Perreau
- Laboratory of AIDS Immunopathogenesis, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Virginie Rozot
- Laboratory of AIDS Immunopathogenesis, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Felicitas Bellutti Enders
- Laboratory of AIDS Immunopathogenesis, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Laboratory of AIDS Immunopathogenesis, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Laboratory of AIDS Immunopathogenesis, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Blankson JN. Effector mechanisms in HIV-1 infected elite controllers: highly active immune responses? Antiviral Res 2009; 85:295-302. [PMID: 19733595 DOI: 10.1016/j.antiviral.2009.08.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 07/30/2009] [Accepted: 08/30/2009] [Indexed: 10/20/2022]
Abstract
Elite controllers (EC) are HIV-1 infected patients control viral replication to a level of <50 copies/ml without antiretroviral therapy. These patients are also known as elite suppressors, or HIV controllers, and they differ from traditional long-term non-progressors (LTNPs) who maintain stable CD4 counts and are asymptomatic without antiretroviral therapy. Recent studies suggest that many EC are infected with replication-competent virus. Thus it appears that host factors such as innate immunity, the humoral immune response, and the cellular immune response are involved in the suppression of viral replication in EC. This article will review the effector mechanisms that are thought to play a role in the remarkable control of viral replication seen in these patients. This article forms part of a special issue of Antiviral Research marking the 25th anniversary of antiretroviral drug discovery and development, Vol 85, issue 1, 2010.
Collapse
Affiliation(s)
- Joel N Blankson
- Broadway Research Bldg, Rm 880, Johns Hopkins University School of Medicine, 722 N. Broadway, Baltimore, MD 21205, United States.
| |
Collapse
|