Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Viruses ; 15(4)2023 04 14.
Article in English | MEDLINE | ID: mdl-37112943

ABSTRACT

During the early 1980s, the first 3 human retroviruses were identified: human T-lymphotropic virus 1 and 2 (HTLV-1 and HTLV-2) and human immunodeficiency virus (HIV) [...].


Subject(s)
Coinfection , HIV Infections , HTLV-I Infections , Human T-lymphotropic virus 1 , Humans , HTLV-I Infections/complications , HIV Infections/complications , Human T-lymphotropic virus 2
2.
Cytometry A ; 103(3): 184-188, 2023 03.
Article in English | MEDLINE | ID: mdl-36470845

ABSTRACT

We developed a highly reproducible 32-marker mass cytometry panel able to measure all canonical immune lineages and perform detailed characterization of both CD4 and CD8 T cells in human peripheral blood mononuclear cells. In this panel, we identify six different T cell memory subsets, as well as markers of activation, cell cycling, and survival. In addition, this panel classifies all major CD4 T cell helper subsets. This panel enables detailed monitoring of CD4 and CD8 T cells in the context of infectious disease, cancer or autoimmunity with limited patient sample use. Detailed methods for standardization and optimization of the panel can be found in Supporting Information.


Subject(s)
CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear , Humans , T-Lymphocyte Subsets , T-Lymphocytes, Helper-Inducer , Lymphocyte Activation , Flow Cytometry/methods , CD4-Positive T-Lymphocytes
3.
Viruses ; 14(11)2022 11 09.
Article in English | MEDLINE | ID: mdl-36366570

ABSTRACT

People living with HIV-1 and HTLV-2 concomitantly show slower CD4+ T cell depletion and AIDS progression, more frequency of the natural control of HIV-1, and lower mortality rates. A similar beneficial effect of this infection has been reported on HCV coinfection reducing transaminases, increasing the spontaneous clearance of HCV infection and delaying the development of hepatic fibrosis. Given the critical role of CD8+ T cells in controlling HIV-1 infection, we analysed the role of CD8+ T cell-mediated cytotoxic activity in coinfected individuals living with HIV-1. One hundred and twenty-eight individuals living with HIV-1 in four groups were studied: two groups with HTLV-2 infection, including individuals with HCV infection (N = 41) and with a sustained virological response (SVR) after HCV treatment (N = 25); and two groups without HTLV-2 infection, including individuals with HCV infection (N = 25) and with a sustained virological response after treatment (N = 37). We found that CD8+ T cell-mediated HIV-1 inhibition in vitro was higher in individuals with HTLV-2. This inhibition activity was associated with a higher frequency of effector memory CD8+ T cells, higher levels of granzyme A and granzyme B cytolytic enzymes, and perforin. Hence, cellular and soluble cytolytic factors may contribute to the lower HIV-1 pre-ART viral load and the HIV-1 proviral load during ART therapy associated with HTLV-2 infection. Herein, we confirmed and expanded previous findings on the role of HTLV-2 in the beneficial effect on the pathogenesis of HIV-1 in coinfected individuals.


Subject(s)
Coinfection , HIV Infections , HIV Seropositivity , HIV-1 , HTLV-II Infections , Hepatitis C , Humans , Human T-lymphotropic virus 2 , HIV-1/physiology , Proviruses , CD8-Positive T-Lymphocytes/pathology , Viral Load , Hepatitis C/complications
4.
Bio Protoc ; 12(6): e4354, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35434196

ABSTRACT

The human immunodeficiency virus (HIV)-1 viral inhibition assay (VIA) measures CD8+ T cell-mediated inhibition of HIV replication in CD4+ T cells and is increasingly used for clinical testing of HIV vaccines and immunotherapies. Different VIAs that differ in length of CD8:CD4 T cell culture periods (6-13 days), purity of CD4 cultures [isolated CD4+ T cells or CD8+ depleted peripheral blood mononuclear cells (PBMCs)], HIV strains (laboratory strains, isolates, reporter viruses) and read-outs of virus inhibition (p24 ELISA, intracellular measurement of p24, luciferase reporter expression, and viral gag RNA) have been reported. Here, we describe multiple modifications to a 7-day VIA protocol, the most impactful being the introduction of independent replicate cultures for both HIV infected-CD4 (HIV-CD4) and HIV-CD4:CD8 T cell cultures. Virus inhibition was quantified using a ratio of weighted averages of p24+ cells in replicate cultures and the corresponding 95% confidence intervals. We identify methodological and analysis changes that could be incorporated into other protocols to improve assay reproducibility. We found that in people living with HIV (PLWH) on antiretroviral therapy (ART), CD8 T cell virus inhibition was largely stable over time, supporting the use of this assay and/or analysis methods to examine therapeutic interventions. Graphic abstract.

6.
Front Immunol ; 12: 666991, 2021.
Article in English | MEDLINE | ID: mdl-34276657

ABSTRACT

The HIV-1 viral inhibition assay (VIA) measures CD8 T cell-mediated inhibition of HIV replication in CD4 T cells and is increasingly used for clinical testing of HIV vaccines and immunotherapies. The VIA has multiple sources of variability arising from in vitro HIV infection and co-culture of two T cell populations. Here, we describe multiple modifications to a 7-day VIA protocol, the most impactful being the introduction of independent replicate cultures for both HIV infected-CD4 (HIV-CD4) and HIV-CD4:CD8 T cell cultures. Virus inhibition was quantified using a ratio of weighted averages of p24+ cells in replicate cultures and the corresponding 95% confidence interval. An Excel template is provided to facilitate calculations. Virus inhibition was higher in people living with HIV suppressed on antiretroviral therapy (n=14, mean: 40.0%, median: 43.8%, range: 8.2 to 73.3%; p < 0.0001, two-tailed, exact Mann-Whitney test) compared to HIV-seronegative donors (n = 21, mean: -13.7%, median: -14.4%, range: -49.9 to 20.9%) and was stable over time (n = 6, mean %COV 9.4%, range 0.9 to 17.3%). Cross-sectional data were used to define 8% inhibition as the threshold to confidently detect specific CD8 T cell activity and determine the minimum number of culture replicates and p24+ cells needed to have 90% statistical power to detect this threshold. Last, we note that, in HIV seronegative donors, the addition of CD8 T cells to HIV infected CD4 T cells consistently increased HIV replication, though the level of increase varied markedly between donors. This co-culture effect may contribute to the weak correlations observed between CD8 T cell VIA and other measures of HIV-specific CD8 T cell function.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Seropositivity/immunology , HIV-1/immunology , Host Microbial Interactions/immunology , Virus Replication/immunology , Antiviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Cells, Cultured , Coculture Techniques , Cross-Sectional Studies , HIV Core Protein p24/immunology , HIV Seropositivity/blood , HIV Seropositivity/drug therapy , HIV Seropositivity/virology , Humans , Treatment Outcome
7.
J Acquir Immune Defic Syndr ; 85(2): 252-256, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32658127

ABSTRACT

OBJECTIVES: This study aims to analyze the association of the presence of common polymorphisms [single nucleotide polymorphisms (SNPs)] on Toll-like receptors (TLRs), such as TLR9-1635A/G, TLR2-1892A/C, TLR2-2258G/A, TLR4-899A/G, and TLR4-1196C/T, with the viral rebound after stopping antiretroviral treatment (ART). CCR5-Δ32 deletion and HLA-A/HLA-B alleles were also analyzed. DESIGN: Interruption of ART may be required to investigate the outcome of strategies aimed to achieve drug-free HIV remission or cure. However, interruption of ART is currently not indicated. This was a retrospective longitudinal study that included 57 long-term suppressed HIV-1-infected individuals. METHODS: TLR SNPs were detected by real-time polymerase chain reaction (PCR). CCR5-Δ32 was analyzed by conventional PCR and HLA-A and HLA-B alleles by PCR-SSOP Luminex. RESULTS: HIV-1 RNA rebound at week 4 after treatment interruption positively correlated with pre-ART HIV-1 load (P = 0.025). The TLR9-1635AA genotype was independently associated with a higher HIV-1 rebound compared with those with AG + GG genotype (multivariate stepwise regression analysis, P = 0.004). Women had lower HIV-1 RNA load both at rebound and during the 72 weeks of follow-up compared with men (P < 0.05 at all time-points), whereas CD4 nadir and CD4 count set-point were similar according to sex. The pre-ART viral load was independently associated with the viral set-point (P = 0.001), whereas the presence of the HLA-A01 allele (P = 0.027) and the CD4 nadir (P = 0.001) were associated with the CD4 count set-point. CONCLUSIONS: The association of the TLR9-1635AA genotype with a higher HIV-1 rebound suggests that this SNP may affect the results from strategies requiring interruption of ART aimed to cure HIV-1 infection.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , Toll-Like Receptor 9/genetics , CD4 Lymphocyte Count , Female , HIV-1 , Humans , Longitudinal Studies , Male , Polymorphism, Single Nucleotide , Retrospective Studies , Viral Load , Virus Latency
9.
Mol Ther Methods Clin Dev ; 15: 9-17, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31534983

ABSTRACT

HIV infection induces a robust T cell response that is sustained by high viremia, but falls following the onset of antiretroviral therapy (ART). Relatively little has been reported on the subsequent stability of the HIV-specific T cell response in individuals on durable therapy. Such data are critical for powering clinical trials testing T cell-based immunotherapies. In a cross-sectional study, HIV-specific T cell responses were detectable by ex vivo interferon (IFN)-γ ELISpot (average ∼1,100 spot-forming units [SFUs]/106 peripheral blood mononuclear cells) in persons living with HIV (PLWH; n = 34), despite median durable ART suppression of 5.0 years. No substantial association was detected between the summed HIV-specific T cell response and the size of the replication-competent HIV reservoir. T cell responses were next measured in participants sampled weekly, monthly, or yearly. HIV-specific T cell responses were highly stable over the time periods examined; within-individual variation ranged from 16% coefficient of variation (CV) for weekly to 27% CV for yearly sampling. These data were used to generate power calculations for future immunotherapy studies. The stability of the HIV-specific T cell response in suppressed PLWH will enable powered studies of small sizes (e.g., n = 6-12), facilitating rapid and iterative testing for T cell-based immunotherapies against HIV.

10.
Nat Biotechnol ; 37(10): 1163-1173, 2019 10.
Article in English | MEDLINE | ID: mdl-31451733

ABSTRACT

A major limitation of current humanized mouse models is that they primarily enable the analysis of human-specific pathogens that infect hematopoietic cells. However, most human pathogens target other cell types, including epithelial, endothelial and mesenchymal cells. Here, we show that implantation of human lung tissue, which contains up to 40 cell types, including nonhematopoietic cells, into immunodeficient mice (lung-only mice) resulted in the development of a highly vascularized lung implant. We demonstrate that emerging and clinically relevant human pathogens such as Middle East respiratory syndrome coronavirus, Zika virus, respiratory syncytial virus and cytomegalovirus replicate in vivo in these lung implants. When incorporated into bone marrow/liver/thymus humanized mice, lung implants are repopulated with autologous human hematopoietic cells. We show robust antigen-specific humoral and T-cell responses following cytomegalovirus infection that control virus replication. Lung-only mice and bone marrow/liver/thymus-lung humanized mice substantially increase the number of human pathogens that can be studied in vivo, facilitating the in vivo testing of therapeutics.


Subject(s)
Coronavirus Infections/virology , Disease Models, Animal , Lung/physiology , Zika Virus Infection/virology , Animals , Antibodies, Viral , Antigen-Presenting Cells , Coronavirus Infections/immunology , Cytokines/genetics , Cytokines/metabolism , Cytomegalovirus/physiology , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Middle East Respiratory Syndrome Coronavirus/immunology , Tropism/immunology , Virus Replication , Zika Virus/immunology , Zika Virus Infection/immunology
SELECTION OF CITATIONS
SEARCH DETAIL