27 research outputs found

    Treatment of two postoperative endophthalmitis cases due to Aspergillus flavus and Scopulariopsis spp. with local and systemic antifungal therapy

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Endophthalmitis is the inflammatory response to invasion of the eye with bacteria or fungi. The incidence of endophthalmitis after cataract surgery varies between 0.072–0.13 percent. Treatment of endophthalmitis with fungal etiology is difficult.</p> <p>Case Presentation</p> <p><b>Case 1: </b>A 71-year old male diabetic patient developed postoperative endophthalmitis due to <it>Aspergillus flavus</it>. The patient was treated with topical amphotericin B ophthalmic solution, intravenous (IV) liposomal amphotericin-B and caspofungin following vitrectomy.</p> <p><b>Case 2: </b>A 72-year old male cachectic patient developed postoperative endophthalmitis due to <it>Scopulariopsis </it>spp. The patient was treated with topical and IV voriconazole and caspofungin.</p> <p>Conclusion</p> <p><it>Aspergillus </it>spp. are responsible of postoperative fungal endophthalmitis. Endophthalmitis caused by <it>Scopulariopsis </it>spp. is a very rare condition. The two cases were successfully treated with local and systemic antifungal therapy.</p

    CD4 is expressed on a heterogeneous subset of hematopoietic progenitors, which persistently harbor CXCR4 and CCR5-tropic HIV proviral genomes in vivo

    Get PDF
    <div><p>Latent HIV infection of long-lived cells is a barrier to viral clearance. Hematopoietic stem and progenitor cells are a heterogeneous population of cells, some of which are long-lived. CXCR4-tropic HIVs infect a broad range of HSPC subtypes, including hematopoietic stem cells, which are multi-potent and long-lived. However, CCR5-tropic HIV infection is limited to more differentiated progenitor cells with life spans that are less well understood. Consistent with emerging data that restricted progenitor cells can be long-lived, we detected persistent HIV in restricted HSPC populations from optimally treated people. Further, genotypic and phenotypic analysis of amplified <i>env</i> alleles from donor samples indicated that both CXCR4- and CCR5-tropic viruses persisted in HSPCs. RNA profiling confirmed expression of HIV receptor RNA in a pattern that was consistent with in vitro and in vivo results. In addition, we characterized a CD4<sup>high</sup> HSPC sub-population that was preferentially targeted by a variety of CXCR4- and CCR5-tropic HIVs in vitro. Finally, we present strong evidence that HIV proviral genomes of both tropisms can be transmitted to CD4-negative daughter cells of multiple lineages in vivo. In some cases, the transmitted proviral genomes contained signature deletions that inactivated the virus, eliminating the possibility that coincidental infection explains the results. These data support a model in which both stem and non-stem cell progenitors serve as persistent reservoirs for CXCR4- and CCR5-tropic HIV proviral genomes that can be passed to daughter cells.</p></div

    Evidence for transmission of proviral genomes from multipotent CD4<sup>+</sup> HSPCs to differentiated peripheral blood cells.

    No full text
    <p>A. Flow cytometric plots showing purity of CD4-negative lineages containing provirus identical to HSPC-derived provirus. “Pre” indicates the cell population post CD4-bead depletion and prior to fluorescence activated cell sorting (FACS). “Post” indicates the cell populations following FACS. Numbers in the upper right corner indicate the frequency of cells in that quadrant. The frequency of CD4<sup>+</sup> cells that were also CD3<sup>+</sup> by gating was 0% (see also <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.t005" target="_blank">Table 5</a>). B and C. Phylogenetic trees showing genetic relationships amongst amplicons. HIV RNA shown is cell-associated (<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.g010" target="_blank">Fig 10B</a>). Arrows indicate location of identical amplicons shown in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.g010" target="_blank">Fig 10</a>. Red lines indicate identical sequences. Scale indicates nucleotide substitutions per site. ACH2, 89.6, BaL, YU-2, HXB2 and NL4-3 are subtype B HIVs. 84ZR085 (84ZR) and 94UG114 (94UG) are subtype D HIV molecular clone outgroups [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.ref032" target="_blank">32</a>]. Phylogenetic analysis was performed by maximum likelihood method using MEGA7[<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.ref033" target="_blank">33</a>] and history was inferred based on the Hasegawa-Kishino-Yano model [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.ref034" target="_blank">34</a>]. The tree with the highest log likelihood is shown. Abbreviations: PBMC, unfractionated peripheral blood mononuclear cells; BMMC, bone marrow mononuclear cell (column flow-through).</p

    Targeting of intermediate progenitors by CCR5-tropic Envs is a conserved property extending to a transmitted/founder virus.

    No full text
    <p>A. Schematic of HIV-7/SF-GFP construct and HIV envelope plasmid used to construct pseudotyped viruses used in C-E. B. Summary table of envelope proteins used to pseudotype HIV-7/SF-GFP virus. C. Representative flow plots of cord blood-derived CD133-sorted cells expanded for four days, transduced with the indicated virus and harvested 3 days post-infection for flow cytometric analysis. In each right panel, GFP<sup>+</sup> cells were overlaid onto plots of the total cell population and the percentage of GFP<sup>+</sup> cells in the CD133<sup>high</sup> and CD133<sup>low</sup> regions is indicated. Gates were determined based on isotype control antibody staining (top panel). D. Summary graph of CD133 MFI for experiments performed as in C. Results are compiled from 11 cord blood experiments. Mean ± standard deviation is indicated; <i>n</i>≥3 for each envelope. 2-tailed Student’s t-test indicates significance for each HIV envelope with respect to VSV-G (**<i>p</i> < 0.01,***<i>p</i> < 0.001, ****<i>p</i> < 0.0001). E. Data from (D) compiled by tropism. Mean ± standard deviation is indicated; one-way ANOVA, <i>p</i> = 0.0002, with Tukey’s Multiple Comparisons Test indicated (**<i>p</i> < 0.01 and ***<i>p</i> < 0.001).</p

    CD4<sup>high</sup> HSPCs include progenitors with multi-lineage potential.

    No full text
    <p>A. Flow cytometric analysis of differentiation markers expressed on bone marrow HSPCs purified as described in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.g001" target="_blank">Fig 1A</a>. For the two right-most panels, numbers indicate percentage of total CD34<sup>+</sup> events in each sort falling into that gate. B. Summary table of frequencies for each phenotypic gate as shown in A. Lineage outputs based upon Doulatov <i>et al</i> [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.ref009" target="_blank">9</a>]. (Abbreviations: HSC, hematopoietic stem cell; MPP, multipotent progenitor; MLP, multilymphoid progenitor; CMP, common myeloid progenitor; MEP, megakaryocyte/erythrocyte progenitor; GMP, granulocyte/monocyte progenitor; B-NK, B and NK cell progenitor; MDC, macrophage and dendritic cell; EMK, erythroid and megakaryocyte) C. Summary graphs depicting the percentage of each subset of the total CD34<sup>+</sup> cells in each sort. Cells were isolated from cord blood (n = 5, circles) or bone marrow (n = 2, squares). For three experiments (2 cord blood and 1 bone marrow), lineage-positive cells were physically or analytically excluded from analysis (open symbols). Mean ± standard deviation is indicated; 2-tailed Student’s t-test (*<i>p</i><0.05, ***<i>p</i><0.001, ****<i>p</i><0.0001).</p

    Sort 2 HSPCs are depleted for HSCs.

    No full text
    <p>A. Diagrammatic representation of HSPC purification. FT, column flow-through; CB, cord blood; BMMC, bone marrow mononuclear cells. B. Representative flow cytometric analysis of HSPC samples from an HIV<sup>+</sup> individual. C. Gates for each HSPC population phenotype and lineage output according to Doulatov <i>et al</i> [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006509#ppat.1006509.ref009" target="_blank">9</a>]. (HSC, hematopoietic stem cell; MPP, multipotent progenitor; MLP, multilymphoid progenitor; CMP, common myeloid progenitor; MEP, megakaryocyte/erythrocyte progenitor; GMP, granulocyte/monocyte progenitor; B-NK, B and NK cell progenitor; MDC, macrophage and dendritic cell; EMK, erythroid and megakaryocyte; G, granulocyte.) D. Representative flow cytometric analysis of differentiation markers expressed on bone marrow HSPCs purified as described in A. For the two right-most panels, numbers indicate percentage of total CD34<sup>+</sup> events from each sort falling into that gate. E. Flow cytometric plot comparing relative numbers of HSCs (CD34<sup>+</sup>CD38<sup>-</sup> cells that are also CD90<sup>+</sup>) in Sort 1 versus Sort 2. F-I. Summary graph showing the relative frequency of the indicated progenitor in each sort, <i>n</i> = 3 uninfected donors. To facilitate comparison, results were normalized to Sort 2 (F and G) or Sort 1 (H—I). Mean ± standard deviation is indicated. J. Summary plots of methylcellulose colony formation assays from three uninfected donors. Mean ± standard deviation is indicated. Colony forming unit (CFU)-E, erythroid; CFU-GM, granulocyte/macrophage and CFU-GEMM, multilineage. (*<i>p</i><0.05 and **<i>p</i><0.01, 2-tailed Student’s t-test).</p
    corecore