8 research outputs found

    Molecular mechanisms of CD8+ T cell mediated control of HIV-1 infection in peripheral blood and lymphoid tissues.

    Get PDF
    Doctoral Degree. University of KwaZulu-Natal, Durban.Naturally induced CD8+ T cells do not clear human immunodeficiency virus (HIV) infection, partly because the virus rapidly escapes CD8+ T cell responses and the effector cells are excluded from HIV reservoirs sites. However, optimizing CD8+ T cell responses could potentially be leveraged in HIV vaccine or cure efforts if epitope escape and barriers to effector CD8+ T cells infiltrating the sites of HIV reservoirs are overcome. In our first study, we described a potential mechanism of HIV-1 control by CD8+ T cells targeting different variants in individuals infected with HIV-1. Our second study focused on describing the molecular regulation of CXCR5 expression in human CD8+ T cells. Study 1 HLA-B*81 is associated with control of HIV-1 subtype C infection, while the closely related allele B*42 is not. Interestingly, both alleles present the immunodominant Gag TL9 epitope, and the magnitude of this response correlates negatively with viral load. To examine the role of T cell receptor (TCR) in this process, we characterized the sequence and function of TL9-specific CD8+ TCR in B*81 and B*42 individuals. TL9-specific CD8+ T cells were identified and isolated using B*81 and/or B*42 TL9 tetramers. TCR beta genes were amplified from single sorted cells and sequenced. Paired alpha genes were identified for selected clones. TCR function was tested using a reporter cell assay where TCR+ Jurkat cells were co-cultured with peptide-pulsed or HIV-1 infected B*81 or B*42 target cells, and signalling quantified by luminescence. TCR recognition was assessed against all single amino acid TL9 variants and results were compared to HIV-1 subtype C sequences. A population of dual-reactive T cells was detected by both B*81- and B*42-TL9 tetramers in 7/9 (78%) B*81 and 4/11 (36%) B*42 individuals; and this population was associated with lower viremia. Mono- and dual-reactive TCR beta sequences were collected from six individuals. In B*81 individuals, all TCRs were highly restricted to TRBV12-3. In B*42 individuals, mono-reactive TCRs encoded a variety of V beta genes, while dual-reactive TCRs were restricted to TRBV12-3 and enriched for public clones. Functional analyses indicated that B*81 TCRs (1 mono, 2 dual) and a dualreactive public B*42 TCR displayed similar TL9 cross-reactivity profiles and enhanced capacity to recognize HIV-1 escape mutations compared to mono-reactive B*42 TCRs. This work highlights the impact of TCR promiscuity on T cell-mediated control of HIV-1. Study 2 HIV-1 infection is difficult to cure even with effective antiretroviral therapy (ART) because of persistent viral replication in immune privileged sites such as the B cell follicles of secondary lymphoid tissues. CD8+ T cells are generally excluded from B cell follicles, partially due to a lack of expression of the follicular homing receptor CXCR5. Recent murine studies have identified CXCR5+ CD8+ T cells, referred to as follicular CD8+ T cells (fCD8s), that localize in B cell follicles. However, the mechanisms governing expression of CXCR5 on human CD8+ T cells are not known. We investigated the epigenetic and transcriptional mechanisms involved in the regulation of CXCR5 expression in human CD8+ T cells. We FACS-sorted CXCR5+CD8+ (fCD8s), CXCR5-CD8+ (non-fCD8s), naĂŻve CD8+ T cells and germinal center T follicular helper cells (GCTfh) from the lymph node of HIV- 1 infected individuals and performed RNA-sequencing (RNA-Seq), DNA methylation assays and the assay for transposase-accessible chromatin using sequencing (ATACSeq). RNA-Seq was used to quantify the expressed genes in FACS-sorted subsets and to determine transcriptional modules governing CXCR5 expression in CD8+ T cells. ATAC-Seq was used to quantify accessible genes, identify the transcriptional factors footprinting and determine epigenetic modules governing CXCR5 expression. DNA methylation, a major epigenetic gene silencing mechanism, was used to profile methylation pattern of the CXCR5 gene region in the sorted subsets. We observed hypermethylation of DNA around the transcriptional start site (TSS) of the CXCR5 gene in non-fCD8s but not in fCD8s. ATAC-Seq analysis revealed a closed chromatin conformation at the TSS in non-fCD8s, but not in fCD8s. Our gene expression data revealed significant differences in the CXCR5 associated factors between GCTfh and fCD8s. Computational analysis further revealed the presence ofa nucleosome at the TSS of fCD8s, which could be a plausible explanation for lower expression of CXCR5 in fCD8s as compared to GCTfh. Together, we identified epigenetic regulations involved in CXCR5 expression in human CD8+ T cells and propose that DNA methylation, chromatin structure and nucleosome positioning cooperatively regulate the expression of CXCR5 in CD8+ T cells. Our data open up the possibility of using epigenetic manipulation as a novel strategy for redirecting CD8+ T cells to B cell follicles where they are needed to eradicate HIV-1 infected cells

    Analysis of viral inhibitory activity of cytotoxic T. Lymphocytes targeting identical epitopes restricted by different class 1 HLA alleles from the same HLA supertype.

    Get PDF
    Master of Medical Sciences in Immunology. University of KwaZulu-Natal, Medical School 2015.Human leukocyte antigen (HLA) polymorphism and the genetic diversity of human immunodeficiency virus (HIV) are the major obstacles for designing an effective HIV Cytotoxic T Lymphocytes (CTLs) based vaccine. Interestingly, recent studies have demonstrated that multiple class I alleles can recognize common epitopes “supertopes” due to the homology of amino acids within the major binding pockets of the peptide binding cleft. The implications of this for vaccine design is that a vaccine containing a small number of highly promiscuous supertopes can confer protection against a wide range of HIV variants. This notion makes supertopes immunogen design an attractive option. However, it is not clear whether supertopes presented in the context of different class I HLA alleles would induce functional equivalent CTL responses. In this study, we investigated the inhibitory activity of CTLs targeting identical epitopes presented by class I HLA alleles from the same superfamily. The viral inhibitory activity was measured using a newly developed CEM-GFP reporter T-cell line (GXR-cell) as target cell. We first compared the inhibitory activity of CTLs from 8 subjects targeting TPQDLNTML (Gag p24 residue 180-188-TL9) epitope presented by HLA-B*81:01 or B*42:01 alleles. We then assessed the inhibitory activity of the 8 subjects’ CTLs when presented with in-vivo occurring mutant (Q182S)-TL9 epitope by HLA-B*81:01 or B*42:01 alleles. Furthermore, we compared the inhibitory activity of CTLs from 4 subjects targeting ISPRTLNAW (Gag p24 residue 147-155-IW9) epitope presented by HLA-B*57:03 or B*58:01 alleles. Comparative analysis of the inhibitory activity of the 8 subjects’ CTLs showed no statistical significant difference when TL9 epitope was presented by HLA-B*81:01 or B*42:01 alleles (1:1; p-value = 0.8785, paired t test), even at low target to effector ratio (1:8; p-value = 0.4418). No statistical significant difference was observed in the inhibitory activity of the 8 subjects’ CTLs when mutant (Q182S)-TL9 epitope was presented by HLA-B*81:01 or B*42:01 alleles (1:1; p-value = 0.8042), same result was observed at low target to effector ratio (1:8; p-value = 0.9396). Comparative analysis of the inhibitory activity of the 4 subjects’ CTLs targeting identical IW9 epitopes presented by HLA-B*57:03 or B*58:01 alleles showed a trend towards significance at target to effector ratio 1:1 (1:1; p-value = 0.0924), but at low target to effector ratio, no significance difference was observed (1:8; p-value = 0.1496). In conclusion, we have demonstrated that there is no observable significant difference in the inhibitory activity of CTLs targeting wildtype TL9 or mutant (Q182S)-TL9 epitopes presented in the context of HLA-B*81:01 or B*42:01 alleles. Thus, TL9 epitope could be immunogenic for individuals expressing HLA-B*81:01 or B*42:01 alleles. We have also shown that the inhibitory activity of CTLs targeting identical IW9 epitopes presented by HLAB* 57:03 or B*58:01 alleles is comparable. Indicating that IW9 epitope could be included in immunogen design for individuals expressing HLA-B*57:03 or B*58:01 alleles. These findings are relevant for HIV vaccine approach that seeks to identify immunogenic supertopes that can be cross-presented in a broadly cross-reactive T cell based vaccine design

    Magnitude and Kinetics of CD8+ T Cell Activation during Hyperacute HIV Infection Impact Viral Set Point

    Get PDF
    CD8[superscript +] T cells contribute to the control of HIV, but it is not clear whether initial immune responses modulate the viral set point. We screened high-risk uninfected women twice a week for plasma HIV RNA and identified 12 hyperacute infections. Onset of viremia elicited a massive HIV-specific CD8[superscript +] T cell response, with limited bystander activation of non-HIV memory CD8[superscript +] T cells. HIV-specific CD8[superscript +] T cells secreted little interferon-Îł, underwent rapid apoptosis, and failed to upregulate the interleukin-7 receptor, known to be important for T cell survival. The rapidity to peak CD8[superscript +] T cell activation and the absolute magnitude of activation induced by the exponential rise in viremia were inversely correlated with set point viremia. These data indicate that rapid, high magnitude HIV-induced CD8[superscript +] T cell responses are crucial for subsequent immune control of acute infection, which has important implications for HIV vaccine design.Bill & Melinda Gates FoundationCollaboration for AIDS Vaccine DiscoveryWitten Family FoundationDan and Marjorie SullivanUrsula BrunnerGary and Loren CohenMark and Lisa Schwartz Foundation,International AIDS Vaccine Initiative (UKZNRSA1001)National Institute of Allergy and Infectious Diseases (U.S.) (R37AI067073)Center for AIDS Research (P30 AI060354

    Analysis of viral inhibitory activity of cytotoxic T lymphocytes targeting identical epitopes restricted by different class I HLA alleles from the same HLA supertype.

    No full text
    M. Med. Sc. University of KwaZulu-Natal, Durban 2015.Abstract available in PDF file

    Dual HLA B*42 and B*81-reactive T cell receptors recognize more diverse HIV-1 Gag escape variants

    Get PDF
    Closely related HLA alleles presenting similar HIV-1 epitopes can be associated with variable clinical outcome. Here the authors report their findings on CD8+ T cell responses to the HIV-1 Gag-p24 TL9 immunodominant epitope in the context of closely related protective and less protective HLA alleles, and their differential effect on viral contro

    Augmentation of HIV-specific T cell function by immediate treatment of hyperacute HIV-1 infection

    No full text
    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.National Institute of Health (U.S.) (5U24AI118672)National Institute of Health (U.S.) (1U54CA217377)National Institute of Health (U.S.) (1R33CA202820)National Institute of Health (U.S.) (2U19AI089992)National Institute of Health (U.S.) (1R01HL134539)National Institute of Health (U.S.) (2RM1HG006193)National Institute of Health (U.S.) (2R01HL095791)National Institute of Health (U.S.) (P01AI039671)Bill and Melinda Gates Foundation (OPP1139972
    corecore