8 research outputs found

    Evidence of <em>Bacteroides fragilis</em> Protection from <em>Bartonella henselae</em>-Induced Damage

    Get PDF
    <div><p><em>Bartonella henselae</em> is able to internalize endothelial progenitor cells (EPCs), which are resistant to the infection of other common pathogens. <em>Bacteroides fragilis</em> is a gram-negative anaerobe belonging to the gut microflora. It protects from experimental colitis induced by <em>Helicobacter hepaticus</em> through the polysaccharide A (PSA). The aim of our study was to establish: 1) whether <em>B. fragilis</em> colonization could protect from <em>B. henselae</em> infection; if this event may have beneficial effects on EPCs, vascular system and tissues. Our <em>in vitro</em> results establish for the first time that <em>B. fragilis</em> can internalize EPCs and competes with <em>B. henselae</em> during coinfection. We observed a marked activation of the inflammatory response by Real-time PCR and ELISA in coinfected cells compared to <em>B. henselae-</em>infected cells (63 <em>vs</em> 23 up-regulated genes), and after EPCs infection with mutant <em>B. fragilis</em> ΔPSA (≅90% up-regulated genes) compared to <em>B. fragilis</em>. Interestingly, in a mouse model of coinfection, morphological and ultrastructural analyses by hematoxylin-eosin staining and electron microscopy on murine tissues revealed that damages induced by <em>B. henselae</em> can be prevented in the coinfection with <em>B. fragilis</em> but not with its mutant <em>B. fragilis</em> ΔPSA. Moreover, immunohistochemistry analysis with anti-Bartonella showed that the number of positive cells per field decreased of at least 50% in the liver (20±4 <em>vs</em> 50±8), aorta (5±1 <em>vs</em> 10±2) and spleen (25±3 <em>vs</em> 40±6) sections of mice coinfected compared to mice infected only with <em>B. henselae</em>. This decrease was less evident in the coinfection with ΔPSA strain (35±6 in the liver, 5±1 in the aorta and 30±5 in the spleen). Finally, <em>B. fragilis</em> colonization was also able to restore the EPC decrease observed in mice infected with <em>B. henselae</em> (0.65 <em>vs</em> 0.06 media). Thus, our data establish that <em>B. fragilis</em> colonization is able to prevent <em>B. henselae</em> damages through PSA.</p> </div

    Morphological analysis of murine coinfected tissue by hematoxylin-eosin staining. A.

    No full text
    <p>Representative microscope images of hematoxylin-eosin staining of liver tissues from each group of mice uninfected, infected with <i>B. henselae</i>, <i>B. fragilis</i> and <i>B. fragilis</i> ΔPSA or coinfected, as detailed. Granulomatous inflammatory infiltrates are predominantly evident in the group of mice infected with <i>B. henselae</i> compared to the group infected with <i>B. henselae</i> and <i>B. fragilis</i> ΔPSA and particularly in the mice coinfected with <i>B. henselae</i> and <i>B. fragilis</i>. Uninfected controls are negative, as well as <i>B. fragilis</i> and <i>B. fragilis</i> ΔPSA groups. <b>B.</b> Representative sections stained with hematoxylin-eosin of aortas from all mice groups as described. In the group of mice infected with <i>B. henselae</i>, minimal lesions are observed in the aorta compared to liver tissues.</p

    <i>B. fragilis</i> and <i>B. henselae</i> internalize EPCs. A.

    No full text
    <p>Confocal images of human early EPCs infected with <i>B. henselae, B. fragilis</i> and <i>B. fragilis</i> ΔPSA at 100 MOI. Cells were stained with anti-<i>Bacteroides</i> (red) or with anti-<i>Bartonella</i> (green) specific antibodies after 24 h from infection. <b>B.</b> EPCs coinfected with <i>B. henselae,</i> and <i>B. fragilis</i> or <i>B. fragilis</i> ΔPSA as indicated. A MOI of 100 was used for all bacteria strains. Cells were stained with anti-<i>Bacteroides</i> (red) and with anti-<i>Bartonella</i> (green) specific antibodies after 24 h from infection.</p

    Ultrastructural analysis of murine infected tissue.

    No full text
    <p>Electron microscopy of liver and aorta: <b>A.</b> Portal triad of normal liver. <b>B.</b> Granuloma around a centrilobular vein of the liver infected with <i>B. henselae</i> with a characteristic neutrophil. <b>C.</b> Normal aortic intima and media. <b>D.</b> White cells in the tunica adventitia of aorta infected with <i>B. henselae</i>. <b>E.</b> Centrilobular vein of liver coinfected with <i>B. henselae</i> and <i>B. fragilis</i>. <b>F.</b> Granuloma in liver coinfected with <i>B. henselae</i> and <i>B. fragilis</i> ΔPSA. <b>G.</b> Endothelium bulging of aorta coinfected with <i>B. henselae</i> and <i>B. fragilis</i>. <b>H.</b> Endothelium bulging of aorta coinfected with <i>B. henselae</i> and <i>B. fragilis</i> ΔPSA. <b>I.</b> Centrilobular vein of liver infected with <i>B. fragilis</i>. <b>J.</b> Centrilobular vein of liver infected with <i>B. fragilis</i> ΔPSA. <b>K.</b> Endothelium of aorta infected with <i>B. fragilis</i>. <b>L.</b> Endothelium of aorta infected with <i>B. fragilis</i> ΔPSA.</p

    Expression levels of inflammatory genes in infected EPCs. A.

    No full text
    <p>Expression levels of 84 inflammatory genes in EPCs infected with <i>B. henselae, B. fragilis, B. fragilis</i> ΔPSA, <i>B. henselae</i> and <i>B. fragilis, B. henselae</i> and <i>B. fragilis</i> ΔPSA. Differential expression data are evaluated <i>vs</i> uninfected EPCs. <b>B.</b> Bar graph showing the expression of some relevant differentially expressed genes encoding chemokines, cytokines and their receptors in <i>B. fragilis- vs B. fragilis</i> ΔPSA-infected EPCs. Data are shown as relative expression levels (<i>B. fragilis</i> ΔPSA-infected cells = 1). <b>C.</b> and <b>D.</b> Expression levels of representative pro-inflammatory (C) and anti-inflammatory (D) genes highly differentially expressed during coinfection of EPCs, in presence and in absence of PSA. <b>E.</b> and <b>F.</b> Absolute quantification of relevant secreted inflammatory proteins detected by ELISA in the EPCs infected with <i>B. henselae,</i> and coinfected with <i>B. fragilis</i> and <i>B. fragilis</i> ΔPSA.</p

    Morphological analysis of murine coinfected tissue by immunohistochemistry. A.

    No full text
    <p>Representative images of immunohistochemistry analysis of murine liver samples with an antibody against <i>B. henselae</i> are shown (206X magnification). Bar graphs show the mean of the positive cell number per field (HPF) as indicated. 20 fields per each section were analyzed and bars represent standard deviations. <b>B.</b> Immunohistochemistry analysis of murine aorta samples.</p

    Murine infection with <i>B. henselae</i> and <i>B. fragilis</i> and detection of bacteria in infected tissues. A.

    No full text
    <p>Scheme of murine infection: three groups of C57BL/6J mice (n = 5) were infected with <i>B. henselae</i>, <i>B. fragilis</i>, <i>B. fragilis</i> ΔPSA respectively and two groups of animals (n = 5) were coinfected with <i>B. henselae</i> and <i>B. fragilis</i> or <i>B. fragilis</i> ΔPSA. <b>B.</b> TEM micrographs of the liver of C57BL/6J mice infected with <i>B. fragilis</i> and <i>B. fragilis</i> ΔPSA respectively, <i>B. henselae</i> and coinfected. <b>C.</b> Immunofluorescence analysis of the livers of animals infected with <i>B. henselae</i>, <i>B. fragilis</i> and coinfected. Livers were sectioned and treated with an anti-<i>B. henselae</i> and a <i>Bacteroides</i> LPS primary antibody followed by TRITC- or FITC-conjugated secondary antibodies respectively. Liver sections from uninfected mice were used as controls (data not shown). Selected merge images of coinfected mice were acquired by confocal microscopy (Magnification: all, X 270). <b>D.</b> Detection of <i>B. henselae gltA</i> gene and <i>B. fragilis frdA</i> gene by nested-PCR from the liver samples of experimentally infected C57BL/6J with <i>B. fragilis</i> and <i>B. fragilis</i> ΔPSA (lane 1–2), <i>B. henselae</i> (lane 3–4), coinfected (lane 5–8), respectively. Genomic DNA extracted from <i>B. henselae</i> and <i>B. fragilis</i> was used as positive control (lane 9).</p
    corecore