15 research outputs found

    KIR3DL01 upregulation on gut natural killer cells in response to SIV infection of KIR- and MHC class I-defined rhesus macaques

    No full text
    <div><p>Natural killer cells provide an important early defense against viral pathogens and are regulated in part by interactions between highly polymorphic killer-cell immunoglobulin-like receptors (KIRs) on NK cells and their MHC class I ligands on target cells. We previously identified MHC class I ligands for two rhesus macaque KIRs: KIR3DL01 recognizes Mamu-Bw4 molecules and KIR3DL05 recognizes Mamu-A1*002. To determine how these interactions influence NK cell responses, we infected KIR3DL01<sup>+</sup> and KIR3DL05<sup>+</sup> macaques with and without defined ligands for these receptors with SIV<sub>mac</sub>239, and monitored NK cell responses in peripheral blood and lymphoid tissues. NK cell responses in blood were broadly stimulated, as indicated by rapid increases in the CD16<sup>+</sup> population during acute infection and sustained increases in the CD16<sup>+</sup> and CD16<sup>-</sup>CD56<sup>-</sup> populations during chronic infection. Markers of proliferation (Ki-67), activation (CD69 & HLA-DR) and antiviral activity (CD107a & TNFĪ±) were also widely expressed, but began to diverge during chronic infection, as reflected by sustained CD107a and TNFĪ± upregulation by KIR3DL01<sup>+</sup>, but not by KIR3DL05<sup>+</sup> NK cells. Significant increases in the frequency of KIR3DL01<sup>+</sup> (but not KIR3DL05<sup>+</sup>) NK cells were also observed in tissues, particularly in the gut-associated lymphoid tissues, where this receptor was preferentially upregulated on CD56<sup>+</sup> and CD16<sup>-</sup>CD56<sup>-</sup> subsets. These results reveal broad NK cell activation and dynamic changes in the phenotypic properties of NK cells in response to SIV infection, including the enrichment of KIR3DL01<sup>+</sup> NK cells in tissues that support high levels of virus replication.</p></div

    NK cell degranulation and cytokine release in response to SIV infection.

    No full text
    <p>Longitudinal changes in the upregulation of CD107a (A & C) and TNFĪ± (B & D) are shown for KIR3DL01<sup>+</sup>, KIR3DL05<sup>+</sup> and KIR3DL01<sup>-</sup>05<sup>-</sup> NK cells in response to stimulation with 721.221 cells (A & B) or without 721.221 cell stimulation (C & D). PBMCs were incubated overnight with or without 721.221 cells in the presence of an antibody to CD107a, and stained the following day with antibodies to differentiate KIR3DL01<sup>+</sup>, KIR3DL05<sup>+</sup> and KIR3DL01<sup>-</sup>05<sup>-</sup> NK cells and for intracellular accumulation of TNFĪ±. Representative gating for CD107a and TNFĪ± staining is shown in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006506#ppat.1006506.s006" target="_blank">S6 Fig</a>. The mean and standard error (error bars) are plotted for the each NK cell population. Significance values for acute (weeks 1ā€“4) and chronic (weeks 6ā€“24) infection compared to pre-infection (week 0) are indicated with asterisks color-coded to the corresponding cell population (p<0.05*, p<0.01**, p< 0.005*** & p<0.001****, mixed effects models).</p

    KIR3DL01 upregulation on NK cells of the gastrointestinal mucosa.

    No full text
    <p>Representative KIR3DL01 versus KIR3DL05 staining on NK cells isolated from colorectal biopsies is shown for two different rhesus macaques before (week -4) and after (week 2) SIV infection. CD16 versus CD56 staining is shown for the KIR3DL01<sup>+</sup> population at week 2 post-infection (right panels).</p

    Longitudinal changes in NK and T cell counts in peripheral blood in response to SIV infection.

    No full text
    <p>Absolute counts for total NK cells (A), CD8<sup>+</sup> T cells (B), CD4<sup>+</sup> T cells (C), KIR3DL01<sup>+</sup> NK cells (D), KIR3DL05<sup>+</sup> NK cells (E), KIR3DL01<sup>-</sup>05<sup>-</sup> NK cells (F), KIR3DL01<sup>+</sup>05<sup>+</sup> NK cells (G), CD16<sup>+</sup> NK cells (H), CD56<sup>+</sup> NK cells (I), and CD16<sup>-</sup>CD56<sup>-</sup> NK cells (J) were monitored by staining whole blood and PBMCs as described in the methods. Individual (K) and geometric mean (L) SIV RNA loads in plasma are shown for Mamu-A1*002<sup>+</sup> (blue) andā€“A1*002<sup>-</sup> (red) animals. Gating strategies for determining absolute lymphocyte counts in blood and the percentages of PBMCs expressing CD16, CD56, KIR3DL01 and KIR3DL05 are shown in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006506#ppat.1006506.s001" target="_blank">S1</a> and <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006506#ppat.1006506.s004" target="_blank">S4</a> Figs. Viral loads were measured using a qRT-PCR assay with a detection threshold of 30 copies/ml (dotted line) and error bars indicate 95% CI for geometric mean values. Statistics were calculated using a mixed effects model by comparing results from acute (week 1ā€“4) and chronic (weeks 6ā€“24) infection to pre-infection (week 0) (p<0.05 *, p<0.01**, p< 0.005*** & p<0.001****).</p

    NK cell proliferation and activation in response to SIV infection.

    No full text
    <p>Longitudinal changes in the expression of Ki-67 (A & B), CD69 (C & D), HLA-DR (E & F), and Ī±4Ī²7 (G & H) were monitored for CD16<sup>+</sup>, CD56<sup>+</sup> and CD16<sup>-</sup>CD56<sup>-</sup> NK cells (left panels: A, C, E & G) and for KIR3DL01<sup>+</sup>, KIR3DL05<sup>+</sup> and KIR3DL01<sup>-</sup>05<sup>-</sup> NK cells (right panels: B, D, F & H). Absolute counts were calculated as a percentage of total NK cell counts by staining PBMCs with antibodies to CD3, CD8, NKG2A, CD16, CD56, KIR3DL01 and KIR3DL05 (tetramer), and to markers of proliferation (Ki-67), activation (CD69 & HLA-DR) and mucosal homing (Ī±4Ī²7). Representative gating for the proliferation and activation panel is shown in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006506#ppat.1006506.s004" target="_blank">S4 Fig</a>. The mean and standard error (error bars) are shown for each NK cell subset. Significance values for acute (weeks 1ā€“4) and chronic (weeks 6ā€“24) infection compared to pre-infection (week 0) are indicated with asterisks color-coded to the corresponding cell population (p<0.05*, p<0.01**, p< 0.005*** & p<0.001****, mixed effects models).</p

    Comparison of the frequencies of KIR3DL01<sup>+</sup> and KIR3DL05<sup>+</sup> NK cells in lymph nodes and gut-associated lymphoid tissues.

    No full text
    <p>The frequencies of KIR3DL05<sup>+</sup> (A), KIR3DL01<sup>+</sup> (B) and KIR3DL01<sup>-</sup>05<sup>-</sup> (C) NK cells in PBMCs, lymph nodes (LN) and gut-associated lymphoid tissues (GALT) were compared prior to SIV infection (week -4) and at weeks 2 and 8 after SIV infection. The frequencies of CD16<sup>+</sup>, CD56<sup>+</sup> and CD16<sup>-</sup>CD56<sup>-</sup> NK cells in lymph nodes (D & E) and gut-associated lymphoid tissues (F & G) expressing KIR3DL05 (D & F) or KIR3DL01 (E & G) were also compared at weeks -4, 2 and 8 pre- and post-infection. Representative gating for differentiating KIR3DL01<sup>+</sup> versus KIR3DL05<sup>+</sup> NK cells in lymphocytes isolated from colorectal biopsies is illustrated in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006506#ppat.1006506.s007" target="_blank">S7 Fig</a>. Significant differences are indicated with asterisks (p<0.05*, p<0.01**, p< 0.005*** & p<0.001****, Mann-Whitney <i>U</i>-test). Samples with less than 30 events per gate were excluded from the analysis.</p

    Frequency and intensity of KIR expression as a function of the presence of MHC class I ligands.

    No full text
    <p>Comparison of the frequency (A) and the mean fluorescence intensity (B) of KIR3DL01 staining on NK cells prior to SIV infection versus the number of MHC class I alleles predicted to encode Mamu-Bw4 ligands for this receptor by linear regression analysis. Comparison of the frequency (C) and the mean fluorescence intensity (D) of KIR3DL05 staining on NK cells for Mamu-A1*002<sup>-</sup>, -A3*13<sup>-</sup> (open circle), Mamu-A1*002<sup>-</sup>, -A3*13<sup>+</sup> (red dot), Mamu-A1*002<sup>+</sup>, -A3*13<sup>-</sup> (black dot), and Mamu-A1*002<sup>+</sup>, -A3*13<sup>+</sup> (black circle, red dot) animals prior to SIV infection by linear regression analysis. Comparison of the frequency (E) and the mean fluorescence intensity (F) of KIR3DL05 staining on NK cells for Mamu-A1*002<sup>+</sup> (blue) versus -A1*002<sup>-</sup> (red) animals at the indicated time points before and after SIV infection. Significant differences are indicated with asterisks (p<0.05* & p<0.01**, Mann-Whitney <i>U</i>-test).</p

    Changes in the frequency of CD16<sup>+</sup>, CD56<sup>+</sup> and CD16<sup>-</sup>CD56<sup>-</sup> NK cells expressing KIR3DL01 and KIR3DL05.

    No full text
    <p>The percentages of CD16<sup>+</sup>, CD56<sup>+</sup> and CD16<sup>-</sup>CD56<sup>-</sup> NK cells that are KIR3DL01<sup>+</sup> (A), KIR3DL05<sup>+</sup> (B) or KIR3DL01<sup>-</sup>05<sup>-</sup> (C) were monitored in peripheral blood. The mean and standard error (error bars) are shown for each subset. Significance values were calculated using a mixed effects model by comparing the results from acute (weeks 1ā€“4) and chronic (weeks 6ā€“24) infection to pre-infection (week 0) and are indicated with asterisks color-coded according to the corresponding NK cell population (p<0.05*, p<0.01**, p< 0.005*** & p<0.001****).</p

    Microbial Translocation and Inflammation Occur in Hyperacute Immunodeficiency Virus Infection and Compromise Host Control of Virus Replication

    No full text
    <div><p>Within the first three weeks of human immunodeficiency virus (HIV) infection, virus replication peaks in peripheral blood. Despite the critical, causal role of virus replication in determining transmissibility and kinetics of progression to acquired immune deficiency syndrome (AIDS), there is limited understanding of the conditions required to transform the small localized transmitted founder virus population into a large and heterogeneous systemic infection. Here we show that during the hyperacute ā€œpre-peakā€ phase of simian immunodeficiency virus (SIV) infection in macaques, high levels of microbial DNA transiently translocate into peripheral blood. This, heretofore unappreciated, hyperacute-phase microbial translocation was accompanied by sustained reduction of lipopolysaccharide (LPS)-specific antibody titer, intestinal permeability, increased abundance of CD4+CCR5+ T cell targets of virus replication, and T cell activation. To test whether increasing gastrointestinal permeability to cause microbial translocation would amplify viremia, we treated two SIV-infected macaque ā€˜elite controllersā€™ with a short-course of dextran sulfate sodium (DSS)ā€“stimulating a transient increase in microbial translocation and a prolonged recrudescent viremia. Altogether, our data implicates translocating microbes as amplifiers of immunodeficiency virus replication that effectively undermine the hostā€™s capacity to contain infection.</p></div
    corecore