12 research outputs found

    MiR-155 Induction by F. novicida but Not the Virulent F. tularensis Results in SHIP Down-Regulation and Enhanced Pro-Inflammatory Cytokine Response

    Get PDF
    The intracellular Gram-negative bacterium Francisella tularensis causes the disease tularemia and is known for its ability to subvert host immune responses. Previous work from our laboratory identified the PI3K/Akt pathway and SHIP as critical modulators of host resistance to Francisella. Here, we show that SHIP expression is strongly down-regulated in monocytes and macrophages following infection with F. tularensis novicida (F.n.). To account for this negative regulation we explored the possibility that microRNAs (miRs) that target SHIP may be induced during infection. There is one miR that is predicted to target SHIP, miR-155. We tested for induction and found that F.n. induced miR-155 both in primary monocytes/macrophages and in vivo. Using luciferase reporter assays we confirmed that miR-155 led to down-regulation of SHIP, showing that it specifically targets the SHIP 3′UTR. Further experiments showed that miR-155 and BIC, the gene that encodes miR-155, were induced as early as four hours post-infection in primary human monocytes. This expression was dependent on TLR2/MyD88 and did not require inflammasome activation. Importantly, miR-155 positively regulated pro-inflammatory cytokine release in human monocytes infected with Francisella. In sharp contrast, we found that the highly virulent type A SCHU S4 strain of Francisella tularensis (F.t.) led to a significantly lower miR-155 response than the less virulent F.n. Hence, F.n. induces miR-155 expression and leads to down-regulation of SHIP, resulting in enhanced pro-inflammatory responses. However, impaired miR-155 induction by SCHU S4 may help explain the lack of both SHIP down-regulation and pro-inflammatory response and may account for the virulence of Type A Francisella

    Detection of microRNA Expression in Human Peripheral Blood Microvesicles

    Get PDF
    MicroRNAs (miRNA) are small non-coding RNAs that regulate translation of mRNA and protein. Loss or enhanced expression of miRNAs is associated with several diseases, including cancer. However, the identification of circulating miRNA in healthy donors is not well characterized. Microvesicles, also known as exosomes or microparticles, circulate in the peripheral blood and can stimulate cellular signaling. In this study, we hypothesized that under normal healthy conditions, microvesicles contain miRNAs, contributing to biological homeostasis.Microvesicles were isolated from the plasma of normal healthy individuals. RNA was isolated from both the microvesicles and matched mononuclear cells and profiled for 420 known mature miRNAs by real-time PCR. Hierarchical clustering of the data sets indicated significant differences in miRNA expression between peripheral blood mononuclear cells (PBMC) and plasma microvesicles. We observed 71 miRNAs co-expressed between microvesicles and PBMC. Notably, we found 33 and 4 significantly differentially expressed miRNAs in the plasma microvesicles and mononuclear cells, respectively. Prediction of the gene targets and associated biological pathways regulated by the detected miRNAs was performed. The majority of the miRNAs expressed in the microvesicles from the blood were predicted to regulate cellular differentiation of blood cells and metabolic pathways. Interestingly, a select few miRNAs were also predicted to be important modulators of immune function.This study is the first to identify and define miRNA expression in circulating plasma microvesicles of normal subjects. The data generated from this study provides a basis for future studies to determine the predictive role of peripheral blood miRNA signatures in human disease and will enable the definition of the biological processes regulated by these miRNA

    SHIP expression is down-regulated in response to <i>F.n.</i> infection.

    No full text
    <p>(A) PBM were infected with <i>F.n.</i> at an MOI of 50 for 24 hours. Cell lysates were analyzed by Western blotting use an anti-SHIP antibody in the top panel. ‘R’ designates resting/uninfected cells and ‘24 h’ designates infected with <i>F.n.</i> The lower panel is a reprobe of the same membrane with an anti-actin antibody. (B) THP-1 were infected with <i>F.n.</i> at an MOI of 100 for 24 hours. Cell lysates were resolved as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g001" target="_blank">Figure 1A</a>. (C) Western blots for SHIP in murine bone marrow-derived macrophages infected with <i>F.n.</i> at an MOI of 50 for 24 hours. Cell lysates were resolved as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g001" target="_blank">Figure 1A</a>. (D) Predicted interaction between miR-155 and the 3′UTR of SHIP (INPP5D) mRNA. (E) Normalized luciferase activity in cells transfected with the 3′UTR of SHIP (psiCHECK_INPP5D) or with vector alone (psiCHECK), and cotransfected with a control <i>Renilla</i> luciferase vector. Synthetic miR-155 or non-specific (scrambled) miRs were subsequently transfected at concentrations of 0, 10, 25 and 50 nM. Luminometer readings were taken 48 hours post-transfection. The graph represents <i>f-luc</i> expression normalized to <i>r-luc</i> expression, then normalized to percent maximal response.</p

    Virulent <i>F.t.</i> elicits suboptimal miR-155 and pro-inflammatory cytokine responses.

    No full text
    <p>(A–B) PBM (n = 7) were infected with <i>F.n.</i> or <i>F.t.</i> at an MOI of 100 for 24 hours. Relative miR-155 (A) and <i>BIC</i> (B) expression were measured by qRT-PCR. (C) PBM were infected with <i>F.n.</i> or <i>F.t.</i> at an MOI of 50 for 24 hours, then cell lysates probed for SHIP by Western blotting (top panel). The lower panel is a reprobe of the same membrane with anti-actin antibody. (D–E) PBM were infected with <i>F.n.</i> or <i>F.t.</i> at an MOI of 50 for 24 hours. TNFα (D) and (E) IL-6 in supernatants were measured by ELISA. Graphs represent the mean and standard deviation of samples from three independent infections. Data were analyzed by a paired Student <i>t</i> test. An asterisk (*) indicates a <i>p</i>-value<0.05.</p

    miR-155 promotes pro-inflammatory cytokine production during <i>F.n.</i> infection.

    No full text
    <p>(A) PBM were transfected with vector control or miR-155 overexpression construct. 22 hours post-transfection, RNA was extracted and miR-155 over-expression was verified by qRT-PCR. (B) SHIP expression was assayed from the samples in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g005" target="_blank">Figure 5A</a>. (C–D) 14 hours post-transfection PBM were infected with <i>F.n.</i> at an MOI of 50 for 8 hours and (C) TNFα and (D) IL-6 measured in supernatants. (E) Wild-type and miR-155<sup>−/−</sup> macrophages were assayed for miR-155 expression. (F) SHIP expression was assayed by qRT-PCR from the samples in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g005" target="_blank">Figure 5E</a>. (G) Wild-type and miR-155<sup>−/−</sup> macrophages were infected with <i>F.n.</i> at an MOI of 50 for eight hours. IL-6 in the supernatant was measured by ELISA. All experiments were performed in triplicate. Graphs represent the mean and standard deviation of samples from three independent experiments. Data were analyzed by a paired Student <i>t</i> test. An asterisk (*) indicates a <i>p</i>-value<0.05.</p

    Model of miR-155 induction and function during <i>Francisella</i> infection.

    No full text
    <p><i>Francisella</i> is recognized on the host cell surface by TLR2. The signal is transmitted through the adaptor protein MyD88. Subsequently MAPKs, PI3K and Akt are activated, which leads to enhanced NFκB activity, inflammatory cytokine production, and effective host response. SHIP negatively regulates the activation of Akt to prevent effective host response. During <i>Francisella</i> infection miR-155 is induced through the TLR signaling pathway, PI3K/Akt, ERK, JNK, and NFκB. MiR-155 induction in turn down-regulates SHIP to promote the activation of the PI3K/Akt pathway and inflammatory cytokine production. The highly virulent <i>F. tularensis</i> suppresses or subverts the induction of miR-155 in human monocytes, while the relatively avirulent <i>F. novicida</i> does not.</p

    Effects of host-cell entry and inflammasome activation on miR-155 induction.

    No full text
    <p>(A) PBM were pretreated for 30 minutes with 5 µg/ml cytochalasin D (CytoD), infected with <i>F.n.</i> at an MOI of 50 for 6 hours, then relative miR-155 expression measured by qRT-PCR. (B) CFU assays were conducted in parallel with the samples from <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g004" target="_blank">Figure 4A</a>. (C) Wild-type (WT) and caspase-1<sup>−/−</sup> bone marrow-derived macrophages were infected with <i>F.n.</i> at an MOI of 50 for 8 hours and miR-155 was assayed by qRT-PCR. (D) ELISAs were done to measure IL-1β in supernatants from the samples in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g004" target="_blank">Figure 4C</a>. Graphs represent the mean and standard deviation from three independent infections. Data were analyzed by a paired Student <i>t</i> test. An asterisk (*) indicates a <i>p</i>-value<0.05.</p

    Bacterial viability contributes to miR-155 induction through the TLR pathway.

    No full text
    <p>(A) PBM were infected with live, heat-killed (HK), or paraformaldehyde-fixed (PFA) <i>F.n.</i> at an MOI of 50 for 24 hours, then miR-155 measured by qRT-PCR. (B) PBM were infected with live <i>F.n.</i> or heat-killed <i>F.n.</i> at an MOI of 50, then with or without gentamicin after 2 hours, then miR-155 was assayed by qRT-PCR. (C) Wild-type (WT) or MyD88<sup>−/−</sup> bone marrow-derived macrophages were infected with <i>F.n.</i> at an MOI of 50 for 24 hours, then relative miR-155 expression measured by qRT-PCR and then converted to fold change over uninfected. (D) ELISA for TNFα secretion in supernatants from the samples in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g003" target="_blank">Figure 3C</a>. (E) Wild-type (WT) and TLR2 (TLR2 Mut) signaling mutant bone marrow-derived macrophages were infected with <i>F.n.</i> at an MOI of 50 for eight hours and miR-155 was assayed by qRT-PCR. (F) WT and TLR2 Mut macrophages were infected at an MOI of 50 of <i>F.n.</i> for 5, 20, 40, and 60 minutes. Cell lysates were analyzed by Western blotting for pIKKα, pNFκBp65, and Actin. (G) PBM were pretreated for 30 minutes with inhibitors of PI3K (LY), ERK (UO), JNK (SP), p38 (SB), or with DMSO vehicle control, infected with <i>F.n.</i> at an MOI of 50 for 6 hours, then miR-155 was assayed by qRT-PCR. (H) PBM were pretreated with the IKK inhibitor BAY7085 (BAY) or DMSO vehicle control for 90 minutes, infected with <i>F.n.</i> at an MOI of 50 for 6 hours, then miR-155 was assayed by qRT-PCR. (I) ELISA for TNFα in supernatants from the samples in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0008508#pone-0008508-g003" target="_blank">Figure 3F</a>. All experiments were performed in triplicate, and each experiment performed three times. Graphs represent mean and standard deviation. Data were analyzed by a paired Student <i>t</i> test. An asterisk (*) indicates a <i>p</i>-value<0.05.</p
    corecore