16 research outputs found

    TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection.

    Get PDF
    HIV infection induces phenotypic and functional changes to CD8+ T cells defined by the coordinated upregulation of a series of negative checkpoint receptors that eventually result in T cell exhaustion and failure to control viral replication. We report that effector CD8+ T cells during HIV infection in blood and SIV infection in lymphoid tissue exhibit higher levels of the negative checkpoint receptor TIGIT. Increased frequencies of TIGIT+ and TIGIT+ PD-1+ CD8+ T cells correlated with parameters of HIV and SIV disease progression. TIGIT remained elevated despite viral suppression in those with either pharmacological antiretroviral control or immunologically in elite controllers. HIV and SIV-specific CD8+ T cells were dysfunctional and expressed high levels of TIGIT and PD-1. Ex-vivo single or combinational antibody blockade of TIGIT and/or PD-L1 restored viral-specific CD8+ T cell effector responses. The frequency of TIGIT+ CD4+ T cells correlated with the CD4+ T cell total HIV DNA. These findings identify TIGIT as a novel marker of dysfunctional HIV-specific T cells and suggest TIGIT along with other checkpoint receptors may be novel curative HIV targets to reverse T cell exhaustion

    CEACAM1 regulates TIM-3-mediated tolerance and exhaustion

    Get PDF
    T-cell immunoglobulin domain and mucin domain-3 (TIM-3, also known as HAVCR2) is an activation-induced inhibitory molecule involved in tolerance and shown to induce T-cell exhaustion in chronic viral infection and cancers[superscript 1, 2, 3, 4, 5]. Under some conditions, TIM-3 expression has also been shown to be stimulatory. Considering that TIM-3, like cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1), is being targeted for cancer immunotherapy, it is important to identify the circumstances under which TIM-3 can inhibit and activate T-cell responses. Here we show that TIM-3 is co-expressed and forms a heterodimer with carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1), another well-known molecule expressed on activated T cells and involved in T-cell inhibition[superscript 6, 7, 8, 9, 10]. Biochemical, biophysical and X-ray crystallography studies show that the membrane-distal immunoglobulin-variable (IgV)-like amino-terminal domain of each is crucial to these interactions. The presence of CEACAM1 endows TIM-3 with inhibitory function. CEACAM1 facilitates the maturation and cell surface expression of TIM-3 by forming a heterodimeric interaction in cis through the highly related membrane-distal N-terminal domains of each molecule. CEACAM1 and TIM-3 also bind in trans through their N-terminal domains. Both cis and trans interactions between CEACAM1 and TIM-3 determine the tolerance-inducing function of TIM-3. In a mouse adoptive transfer colitis model, CEACAM1-deficient T cells are hyper-inflammatory with reduced cell surface expression of TIM-3 and regulatory cytokines, and this is restored by T-cell-specific CEACAM1 expression. During chronic viral infection and in a tumour environment, CEACAM1 and TIM-3 mark exhausted T cells. Co-blockade of CEACAM1 and TIM-3 leads to enhancement of anti-tumour immune responses with improved elimination of tumours in mouse colorectal cancer models. Thus, CEACAM1 serves as a heterophilic ligand for TIM-3 that is required for its ability to mediate T-cell inhibition, and this interaction has a crucial role in regulating autoimmunity and anti-tumour immunity.American Association for Cancer Research. Pancreatic Cancer Action Networ

    Engineering modular intracellular protein sensor-actuator devices

    No full text
    Understanding and reshaping cellular behaviors with synthetic gene networks requires the ability to sense and respond to changes in the intracellular environment. Intracellular proteins are involved in almost all cellular processes, and thus can provide important information about changes in cellular conditions such as infections, mutations, or disease states. Here we report the design of a modular platform for intrabody-based protein sensing-actuation devices with transcriptional output triggered by detection of intracellular proteins in mammalian cells. We demonstrate reporter activation response (fluorescence, apoptotic gene) to proteins involved in hepatitis C virus (HCV) infection, human immunodeficiency virus (HIV) infection, and Huntington’s disease, and show sensor-based interference with HIV-1 downregulation of HLA-I in infected T cells. Our method provides a means to link varying cellular conditions with robust control of cellular behavior for scientific and therapeutic applications

    Soluble T Cell Immunoglobulin Mucin Domain 3 Is Shed from CD8+ T Cells by the Sheddase ADAM10, Is Increased in Plasma during Untreated HIV Infection, and Correlates with HIV Disease Progression

    No full text
    UnlabelledChronic HIV infection results in a loss of HIV-specific CD8(+) T cell effector function, termed "exhaustion," which is mediated, in part, by the membrane coinhibitory receptor T cell immunoglobulin mucin domain-3 (Tim-3). Like many other receptors, a soluble form of this protein has been described in human blood plasma. However, soluble Tim-3 (sTim-3) is poorly characterized, and its role in HIV disease is unknown. Here, we show that Tim-3 is shed from the surface of responding CD8(+) T cells by the matrix metalloproteinase ADAM10, producing a soluble form of the coinhibitory receptor. Despite previous reports in the mouse model, no alternatively spliced, soluble form of Tim-3 was observed in humans. Shed sTim-3 was found in human plasma and was significantly elevated during early and chronic untreated HIV infection, but it was not found differentially modulated in highly active antiretroviral therapy (HAART)-treated HIV-infected subjects or in elite controllers compared to HIV-uninfected subjects. Plasma sTim-3 levels were positively correlated with HIV load and negatively correlated with CD4 counts. Thus, plasma sTim-3 shedding correlated with HIV disease progression. Despite these correlations, we found that shedding Tim-3 did not improve the function of CD8(+) T cells in terms of gamma interferon production or prevent their apoptosis through galectin-9. Further characterization studies of sTim-3 function are needed to understand the contribution of sTim-3 in HIV disease pathogenesis, with implications for novel therapeutic interventions.ImportanceDespite the overall success of HAART in slowing the progression to AIDS in HIV-infected subjects, chronic immune activation and T cell exhaustion contribute to the eventual deterioration of the immune system. Understanding these processes will aid in the development of interventions and therapeutics to be used in combination with HAART to slow or reverse this deterioration. Here, we show that a soluble form of T cell exhaustion associated coinhibitory molecule 3, sTim-3, is shed from the surface of T cells. Furthermore, sTim-3 is elevated in the plasma of treatment-naive subjects with acute or chronic HIV infection and is associated with markers of disease progression. This is the first study to characterize sTim-3 in human plasma, its source, and mechanism of production. While it is still unclear whether sTim-3 contributes to HIV pathogenesis, sTim-3 may represent a new correlate of HIV disease progression

    Effect of <i>in vitro</i> blockade with anti-TIGIT/anti-PD-L1 mAbs on HIV-specific CD8<sup>+</sup> T cell responses.

    No full text
    <p><i>Ex vivo</i> PBMCs from chronically HIV-infected individuals were stimulated with HIV Gag peptide pool in the presence of mAb blocking antibodies. (A) Representative flow cytometry plots gated on CD8<sup>+</sup> T cells, showing IFN-Îł responses from an HIV-infected individual. No HIV-1 Gag stimulation with an isotype control, HIV-1 Gag stimulation with an isotype control, HIV-1 Gag stimulation with anti-TIGIT, HIV-1 Gag stimulation with anti-PD-L1, HIV-1 Gag stimulation with dual blockade (anti-TIGIT + anti-PD-L1) and a positive control (anti-CD3 + anti-CD28 Dynabeads). (B) Compiled data showing variation in the frequency (%) of IFN-Îł in responses to HIV-1 Gag peptide pool with isotype control or mAb blockade; TIGIT blockade (left panel), PD-L1 blockade (middle panel), and dual blockade (right panel) (<i>n</i> = 25). P values were calculated by Wilcoxon matched-pairs signed ranked test. (C) Representative flow cytometry plots gated on CD8<sup>+</sup> T cells from HIV-infected individuals, showing intermediate and high CFSE dilution in response to HIV-1 Gag peptide pool stimulation in the presence of either an isotype control, anti-TIGIT mAb, anti-PD-L1 mAb, a combination of both anti-TIGIT and anti-PD-L1 mAbs or anti-CD3 + anti-CD28 Dynabeads as a positive control. (D) Graphs show compiled data showing variation in the frequency (%) of CFSE<sup>dim</sup> in responses to HIV-1 Gag peptide pool with either an isotype control or mAb blockade; TIGIT blockade (left panel), PD-L1 blockade (middle panel), and dual blockade (right panel) (<i>n</i> = 24). P values were calculated by Wilcoxon matched-pairs signed ranked test.</p

    Expression of TIGIT on T cells during HIV infection.

    No full text
    <p>Cryopreserved PBMCs were thawed and surface phenotyped for TIGIT expression. Representative flow cytometry flow plots showing TIGIT expression on (A) CD8<sup>+</sup> or (B) CD4<sup>+</sup> T cells compared to fluorescence minus one (FMO) control. Graphs show compiled data of TIGIT expression on (C) CD8<sup>+</sup> and (D) CD4<sup>+</sup> T cells stratified by disease: HIV-uninfected healthy donors (HD, X; <i>n</i> = 20), acute HIV-infection (AI, open diamond; <i>n</i> = 24), aviremic cART suppressed (AS, open triangles; <i>n</i> = 20), aviremic elite controllers (EC, open squares; <i>n</i> = 20), and chronic HIV viremic non-controllers (NC, open circles; <i>n</i> = 20). P values were calculated using one-way ANOVA, followed by Tukey’s multiple comparisons test. Graphs show correlation of total chronic infected (+: AS, EC, and NC; left panel, <i>n</i> = 60) and non-controllers (right panel, <i>n</i> = 20) frequency (%) of (E) TIGIT<sup>+</sup> CD8<sup>+</sup> and (F) TIGIT<sup>+</sup> CD4<sup>+</sup> T cells against clinical CD4 Count (cells/mm<sup>3</sup>). Graphs show correlation of total chronic infected (+: AS, EC, and NC; left panel, <i>n</i> = 60) and elite controllers (right panel, <i>n</i> = 20) frequency (%) of (G) TIGIT<sup>+</sup> CD8<sup>+</sup> and (H) TIGIT<sup>+</sup> CD4<sup>+</sup> T cells against frequency (%) of T cell activation (CD38<sup>+</sup>HLA-DR<sup>+</sup>). Graphs show correlation of frequency (%) of (I) TIGIT<sup>+</sup> CD8<sup>+</sup> and (J) TIGIT<sup>+</sup> CD4<sup>+</sup> T cell among aviremic HIV infected “ART initiators” with known duration of long-term viral suppression from the SCOPE cohort (L-AS, <i>n</i> = 19, open inverted triangles) versus copies of CD4<sup>+</sup> T cell associated HIV DNA per million CD4<sup>+</sup> T cells (log<sub>10</sub>). Spearman’s rho tests were performed for correlations.</p
    corecore