15 research outputs found

    MyD88-dependent signaling influences fibrosis and alternative macrophage activation during Staphylococcus aureus biofilm infection.

    Get PDF
    Bacterial biofilms represent a significant therapeutic challenge based on their ability to evade host immune and antibiotic-mediated clearance. Recent studies have implicated IL-1β in biofilm containment, whereas Toll-like receptors (TLRs) had no effect. This is intriguing, since both the IL-1 receptor (IL-1R) and most TLRs impinge on MyD88-dependent signaling pathways, yet the role of this key adaptor in modulating the host response to biofilm growth is unknown. Therefore, we examined the course of S. aureus catheter-associated biofilm infection in MyD88 knockout (KO) mice. MyD88 KO animals displayed significantly increased bacterial burdens on catheters and surrounding tissues during early infection, which coincided with enhanced dissemination to the heart and kidney compared to wild type (WT) mice. The expression of several proinflammatory mediators, including IL-6, IFN-γ, and CXCL1 was significantly reduced in MyD88 KO mice, primarily at the later stages of infection. Interestingly, immunofluorescence staining of biofilm-infected tissues revealed increased fibrosis in MyD88 KO mice concomitant with enhanced recruitment of alternatively activated M2 macrophages. Taken in the context of previous studies with IL-1β, TLR2, and TLR9 KO mice, the current report reveals that MyD88 signaling is a major effector pathway regulating fibrosis and macrophage polarization during biofilm formation. Together these findings represent a novel example of the divergence between TLR and MyD88 action in the context of S. aureus biofilm infection

    Neuroinflammation leads to region-dependent alterations in astrocyte gap junction communication and hemichannel activity.

    Get PDF
    Inflammation attenuates gap junction (GJ) communication in cultured astrocytes. Here we used a well-characterized model of experimental brain abscess as a tool to query effects of the CNS inflammatory milieu on astrocyte GJ communication and electrophysiological properties. Whole-cell patch-clamp recordings were performed on green fluorescent protein (GFP)-positive astrocytes in acute brain slices from glial fibrillary acidic protein-GFP mice at 3 or 7 d after Staphylococcus aureus infection in the striatum. Astrocyte GJ communication was significantly attenuated in regions immediately surrounding the abscess margins and progressively increased to levels typical of uninfected brain with increasing distance from the abscess proper. Conversely, astrocytes bordering the abscess demonstrated hemichannel activity as evident by enhanced ethidium bromide (EtBr) uptake that could be blocked by several pharmacological inhibitors, including the connexin 43 (Cx43) mimetic peptide Gap26, carbenoxolone, the pannexin1 (Panx1) mimetic peptide (10)Panx1, and probenecid. However, hemichannel opening was transient with astrocytic EtBr uptake observed near the abscess at day 3 but not day 7 after infection. The region-dependent pattern of hemichannel activity at day 3 directly correlated with increases in Cx43, Cx30, Panx1, and glutamate transporter expression (glial L-glutamate transporter and L-glutamate/L-aspartate transporter) along the abscess margins. Changes in astrocyte resting membrane potential and input conductance correlated with the observed changes in GJ communication and hemichannel activity. Collectively, these findings indicate that astrocyte coupling and electrical properties are most dramatically affected near the primary inflammatory site and reveal an opposing relationship between the open states of GJ channels versus hemichannels during acute infection. This relationship may extend to other CNS diseases typified with an inflammatory component

    MyD88-dependent signaling influences fibrosis and alternative macrophage activation during Staphylococcus aureus biofilm infection.

    Get PDF
    Bacterial biofilms represent a significant therapeutic challenge based on their ability to evade host immune and antibiotic-mediated clearance. Recent studies have implicated IL-1β in biofilm containment, whereas Toll-like receptors (TLRs) had no effect. This is intriguing, since both the IL-1 receptor (IL-1R) and most TLRs impinge on MyD88-dependent signaling pathways, yet the role of this key adaptor in modulating the host response to biofilm growth is unknown. Therefore, we examined the course of S. aureus catheter-associated biofilm infection in MyD88 knockout (KO) mice. MyD88 KO animals displayed significantly increased bacterial burdens on catheters and surrounding tissues during early infection, which coincided with enhanced dissemination to the heart and kidney compared to wild type (WT) mice. The expression of several proinflammatory mediators, including IL-6, IFN-γ, and CXCL1 was significantly reduced in MyD88 KO mice, primarily at the later stages of infection. Interestingly, immunofluorescence staining of biofilm-infected tissues revealed increased fibrosis in MyD88 KO mice concomitant with enhanced recruitment of alternatively activated M2 macrophages. Taken in the context of previous studies with IL-1β, TLR2, and TLR9 KO mice, the current report reveals that MyD88 signaling is a major effector pathway regulating fibrosis and macrophage polarization during biofilm formation. Together these findings represent a novel example of the divergence between TLR and MyD88 action in the context of S. aureus biofilm infection

    MyD88 loss during <i>S. aureus</i> biofilm infection augments alternatively activated M2 macrophage accumulation.

    No full text
    <p>Biofilm infections were established in MyD88 knockout (KO) and wild type (WT) mice following the inoculation of 10<sup>3</sup> CFU of USA300 LAC::<i>lux</i> into the lumen of subcutaneous implanted catheters. Animals were sacrificed at day 7 following <i>S. aureus</i> infection, whereupon tissues surrounding infected catheters were collected to quantitate M2 (A) and M1 (B) macrophage infiltrates by FACS on the basis of CD206 and IRF-5 staining, respectively. Results are expressed as the percent macrophages (F4/80<sup>+</sup>) that were also positive for CD206 or IRF-5 after correction for isotype control staining and represent the mean ± SEM of three independent experiments. Significant differences between MyD88 KO and WT infiltrates are denoted by asterisks (*<i>p</i><0.05).</p

    MyD88-dependent signals influence the host tissue response to <i>S. aureus</i> biofilms.

    No full text
    <p>Biofilm-infected tissues were recovered from MyD88 knockout (KO) and wild type (WT) mice at the indicated time points following infection, whereupon sections were processed by hematoxylin and eosin (H&E) staining to demonstrate changes in tissue architecture (A). The deposition of host material surrounding infected catheters is denoted by arrows. (B) Quantitation of the fibrotic thickness surrounding <i>S. aureus</i> biofilms of MyD88 KO and WT mice. Results represent measurements taken from at least seven individual animals per group for each time point where significant differences are indicated with asterisks (*<i>p</i><0.05).</p

    Macrophage recruitment into <i>S. aureus</i> biofilms is attenuated in MyD88 KO mice during early infection.

    No full text
    <p>Biofilms were established in MyD88 knockout (KO) and wild type (WT) mice following the inoculation of 10<sup>3</sup> CFU of USA300 LAC::<i>lux</i> into the lumen of subcutaneous implanted catheters. Animals were sacrificed at the indicated time points following <i>S. aureus</i> infection, whereupon tissues surrounding infected catheters were collected to quantitate macrophage infiltrates by FACS. Results are expressed as the absolute number of F4/80<sup>+</sup> macrophages after normalization to adjust for the recovery of different cell numbers from MyD88 KO and WT mice and represent the mean ± SEM of three independent experiments. Significant differences in macrophage infiltrates are denoted by an asterisk (*<i>p</i><0.05).</p

    MyD88 loss leads to early impairments in <i>S. aureus</i> containment at the site of biofilm infection.

    No full text
    <p>Biofilm infections were established in MyD88 knockout (KO) and wild type (WT) mice following the inoculation of 10<sup>3</sup> CFU of USA300 LAC::<i>lux</i> into the lumen of subcutaneous implanted catheters. Animals were sacrificed at the indicated days following <i>S. aureus</i> biofilm infection, whereupon the heart and kidneys were removed to quantitate bacterial burdens with results expressed as CFU per mg tissue. Results are presented from individual animals in each group combined from a total of at least 2 independent experiments with bars representing the mean of each group. Significant differences in bacterial burdens between MyD88 KO and WT mice are denoted by asterisks (*<i>p</i><0.05).</p

    Type I collagen deposition surrounding <i>S. aureus</i> biofilms is enhanced following MyD88 loss.

    No full text
    <p>Biofilm-infected tissues were recovered from MyD88 knockout (KO) and wild type (WT) mice at the indicated time points following infection, whereupon sections were processed by immunofluorescence staining for type I collagen (red; A). Tissues were stained with DAPI (blue) to demarcate nuclei and asterisks denote the original location of infected catheters that were non-adherent to glass slides. (B) Quantitation of type I collagen fluorescence surrounding <i>S. aureus</i> biofilms of MyD88 KO and WT mice. Results are representative of tissues collected from five individual animals per group for each time point where significant differences are indicated with an asterisk (*<i>p</i><0.05).</p

    Loss of MyD88-dependent signaling augments the expression of genes associated with alternatively activated M2 macrophages following <i>S. aureus</i> biofilm exposure.

    No full text
    <p>Bone marrow-derived macrophages from MyD88 KO or WT mice were co-cultured with <i>S. aureus</i> biofilms <i>in vitro</i> for 2 h, whereupon viable macrophages were purified by FACS and RNA immediately isolated for qRT-PCR analysis. The expression levels of M1 (A), M2 (B), and extracellular matrix (C) genes in macrophages exposed to <i>S. aureus</i> biofilms were calculated after normalizing signals against GAPDH and are presented as fold-change relative to unstimulated macrophages. Results represent the mean ± SEM of at least two independent experiments (*<i>p</i><0.05; **<i>p</i><0.001).</p
    corecore