5 research outputs found

    Design, Synthesis, and Evaluation of Novel Hybrid Efflux Pump Inhibitors for Use against Mycobacterium tuberculosis

    No full text
    Efflux pumps are considered a major potential contributor to the development of various forms of resistance in Mycobacterium tuberculosis leading to the emergence of multidrug-resistant tuberculosis (TB). Verapamil (VER) and tricyclic chemosensitizers such as the phenothiazines are known to possess efflux pump inhibition properties and have demonstrated significant efficacy in various TB disease models. Novel hybrid molecules based on fusion of the VER substructure with various tricyclic, as well as nontricyclic, chemosensitizer cores or their structural motifs are described. These hybrid compounds were evaluated in vitro and ex vivo individually for their intrinsic activity and in combination for their potentiating potential with the frontline anti-TB drugs, rifampin and isoniazid. In addition, efflux pump inhibition was assessed in an ethidium bromide assay. This study led to the identification of novel compounds, termed hybrid efflux pump inhibitors, with intrinsic antimycobacterial activities (MIC<sub>90</sub> ≤ 3.17 μg/mL) and intracellular activity in macrophages at a low concentration (≤6.25 μg/mL)

    Proposed model of granzyme A-mediated suppression of intracellular mycobacterial growth by γ<sub>9</sub>δ<sub>2</sub> T cells.

    No full text
    <p>Mycobacteria-infected macrophages present antigens to γ<sub>9</sub>δ<sub>2</sub> T cells (1) which secrete granzyme A upon activation (2). Granzyme A in turn induces TNF-α production by infected and/or bystander macrophages (3) apparently independent of perforin. TNF-α activates intracellular mechanisms which alone or in concert with other unknown granzyme A-induced intracellular mechanisms suppress mycobacterial growth (4).</p

    TNF-α is critical for γ<sub>9</sub>δ<sub>2</sub> T cell-mediated inhibition of intracellular mycobacterial growth.

    No full text
    <p><i>A</i>, γ<sub>9</sub>δ<sub>2</sub> T cell lines significantly suppressed intracellular growth of mycobacteria (n = 5; *p<0.05 by Wilcoxon matched-pairs test). γ<sub>9</sub>δ<sub>2</sub> T cells were co-cultured with BCG-infected macrophages for 3 d. Surviving bacteria were quantified by H<sup>3</sup>-uridine incorporation and compared with cultures absent of γ<sub>9</sub>δ<sub>2</sub> T cells. <i>B</i>, The effector mechanism responsible for γ<sub>9</sub>δ<sub>2</sub> T cell-mediated mycobacterial growth inhibition involves soluble factors. γ<sub>9</sub>δ<sub>2</sub> T cells were stimulated in the top chambers of semi-permeable transwell membrane (0.4 µm pores) systems with BCG-infected macrophages and/or soluble HMB-PP. The levels of intracellular growth inhibition were assayed in the bottom chambers and compared to the levels of inhibition achieved by direct co-culture of γ<sub>9</sub>δ<sub>2</sub> T cells with the bottom layer of BCG-infected macrophages (*p<0.05, n = 5 by Wilcoxon matched pairs tests compared with unstimulated γ<sub>9</sub>δ<sub>2</sub> T cells separated by transwells; **p<0.02, n = 7 by Wilcoxon matched pairs tests compared with HMB-PP stimulated γδ T cells separated by transwells; ***p<0.003, n = 15 by Wilcoxon matched pairs tests compared with HMB-PP stimulated γδ T cells separated by transwells). <i>C</i>, Neutralization of TNF-α alone prevents γ<sub>9</sub>δ<sub>2</sub> T cell-mediated inhibition while neutralization of IFN-γ alone did not (*p<0.02, n = 7 by Wilcoxon matched pairs tests compared with no antibody control; **p<0.02, n = 6 by Wilcoxon matched pairs tests compared with no antibody control; n.s. – not significantly different). γ<sub>9</sub>δ<sub>2</sub> T cells were co-cultured with BCG-infected macrophages in the presence or absence of the indicated neutralizing antibody and the surviving bacteria quantified by H<sup>3</sup>-uridine incorporation. <i>D</i>, shRNA-mediated knockdown of TNF-α in macrophages, but not γ<sub>9</sub>δ<sub>2</sub> T cells, eliminates γ<sub>9</sub>δ<sub>2</sub> T cell-mediated mycobacterial growth inhibition. (*p<0.02, n = 7 by Wilcoxon matched pairs tests compared with adenovirus expressing the empty vector). γ<sub>9</sub>δ<sub>2</sub> T cells were co-cultured with BCG-infected macrophages. Prior to co-culture, either the T cells or uninfected macrophages were infected with shRNA expressing Adenovirus. Surviving bacteria were quantified by H<sup>3</sup>-uridine incorporation.</p

    Granzyme A secretion by γ<sub>9</sub>δ<sub>2</sub> T cells mediates inhibition of intracellular mycobacteria by induction of inflammatory responses in mycobacteria-infected macrophages.

    No full text
    <p><i>A</i>, The levels of granzyme A produced by γ<sub>9</sub>δ<sub>2</sub> T cells are directly and highly correlated with inhibition of intracellular mycobacterial growth (r = 0.7564; p<0.0001). Supernatant levels of granzyme A, measured by CBA, were correlated with the level of mycobacterial growth inhibition observed in γ<sub>9</sub>δ<sub>2</sub> T cell co-cultures with BCG-infected macrophages. <i>B</i>, Purified granzyme A induces the production of pro-inflammatory cytokines TNF-α and IL-1β by BCG-infected macrophages (*p<0.05, n = 4; statistical comparisons performed using Friedman's test). The indicated concentrations of purified granzyme A were added to BCG-infected monocytes for 3 days and the levels of TNF-α and IL-1β in the culture supernatants determined by CBA. <i>C-D</i>, Purified granzyme A alone can induce inhibition of intracellular mycobacterial growth in the absence of perforin. The indicated concentrations of granzyme A were added to BCG-infected (<i>C</i>) or <i>M. tuberculosis</i> H37Rv-infected (<i>D</i>) co-cultures for 3 days and the surviving bacteria quantified by H<sup>3</sup>-uridine incorporation and CFU counting. (*p<0.03; n = 6–12 by Wilcoxon matched pairs test comparing levels of mycobacterial intracellular growth in cultures with and without added purified granzyme A). Purified granzyme A had no direct effects on extracellular mycobacterial growth (data not shown). <i>E</i>, siRNA knockdown of granzyme A in γ<sub>9</sub>δ<sub>2</sub> T cells reduces the inhibitory activity of γ<sub>9</sub>δ<sub>2</sub> T cells for mycobacterial growth (*p<0.05; n = 3 by one-way ANOVA comparing levels of intracellular mycobacterial growth inhibition in cultures with or without γ<sub>9</sub>δ<sub>2</sub> T cells producing granzyme A). γ<sub>9</sub>δ<sub>2</sub> T cells transduced with the indicated siRNA-lentivirus targeting GzmA or a negative control (NC) were co-cultured with BCG-infected macrophages for 3 days and surviving bacteria quantified by H<sup>3</sup>-uridine incorporation.</p

    Classical effector mechanisms involving direct contact of γ<sub>9</sub>δ<sub>2</sub> T cells with BCG-infected macrophages are not required for mycobacterial suppression.

    No full text
    <p>γ<sub>9</sub>δ<sub>2</sub> T cells were co-cultured with BCG-infected macrophages in the presence or absence of the indicated treatment and surviving bacteria quantified by H<sup>3</sup>-uridine incorporation. Blocking of Fas/FasL interactions did not prevent γ<sub>9</sub>δ<sub>2</sub> T cell-mediated inhibition (p = 0.23, n = 7). In contrast, γ<sub>9</sub>δ<sub>2</sub> T cells pre-treated for 16 hours with 25 mM strontium chloride were unable to mediate mycobacterial inhibition (*p<0.05, n = 5 by Wilcoxon matched pairs test compared with untreated γ<sub>9</sub>δ<sub>2</sub> T cells). Despite strontium depletion indicating the importance of γ<sub>9</sub>δ<sub>2</sub> T cell cytolytic granules for mycobacterial inhibitory effects, antibody neutralization of perforin had no effect (p = 0.28, n = 3).</p
    corecore