16 research outputs found

    Bilayer Metasurfaces for Dual- and Broadband Optical Antireflection

    No full text
    Optical antireflection has long been pursued for a wide range of applications, but existing approaches encounter issues in the performance, bandwidth, and structure complexity, particularly in the long-wavelength infrared regime. Here we present the demonstration of bilayer metasurfaces that accomplish dual- and broadband optical antireflection in the terahertz and mid-infrared spectral ranges. By simply tailoring the structural geometry and dimensions, we show that subwavelength metal/dielectric structures enable dramatic reduction of Fresnel reflection and significant enhancement of transmission at a substrate surface, operating either at two discrete narrow bands or over a broad bandwidth up to 28%. We also use a semianalytical interference model to interpret the obtained results, in which we find that the dispersion of the constituent structures plays a critical role in achieving the observed broadband optical antireflection

    PPARĪ³ is critical for <i>Mycobacterium tuberculosis</i> induction of Mcl-1 and limitation of human macrophage apoptosis

    No full text
    <div><p>Peroxisome proliferator-activated receptor (PPAR)Ī³ is a global transcriptional regulator associated with anti-inflammatory actions. It is highly expressed in alveolar macrophages (AMs), which are unable to clear the intracellular pathogen <i>Mycobacterium tuberculosis (M</i>.<i>tb)</i>. Although <i>M</i>.<i>tb</i> infection induces PPARĪ³ in human macrophages, which contributes to <i>M</i>.<i>tb</i> growth, the mechanisms underlying this are largely unknown. We undertook NanoString gene expression analysis to identify novel PPARĪ³ effectors that condition macrophages to be more susceptible to <i>M</i>.<i>tb</i> infection. This revealed several genes that are differentially regulated in response to PPARĪ³ silencing during <i>M</i>.<i>tb</i> infection, including the Bcl-2 family members Bax (pro-apoptotic) and Mcl-1 (pro-survival). Apoptosis is an important defense mechanism that prevents the growth of intracellular microbes, including <i>M</i>.<i>tb</i>, but is limited by virulent <i>M</i>.<i>tb</i>. This suggested that <i>M</i>.<i>tb</i> differentially regulates Mcl-1 and Bax expression through PPARĪ³ to limit apoptosis. In support of this, gene and protein expression analysis revealed that Mcl-1 expression is driven by PPARĪ³ during <i>M</i>.<i>tb</i> infection in human macrophages. Further, 15-lipoxygenase (15-LOX) is critical for PPARĪ³ activity and Mcl-1 expression. We also determined that PPARĪ³ and 15-LOX regulate macrophage apoptosis during <i>M</i>.<i>tb</i> infection, and that pre-clinical therapeutics that inhibit Mcl-1 activity significantly limit <i>M</i>.<i>tb</i> intracellular growth in both human macrophages and an <i>in vitro</i> TB granuloma model. In conclusion, identification of the novel PPARĪ³ effector Mcl-1 has determined PPARĪ³ and 15-LOX are critical regulators of apoptosis during <i>M</i>.<i>tb</i> infection and new potential targets for host-directed therapy for <i>M</i>.<i>tb</i>.</p></div

    Model.

    No full text
    <p>In human macrophages, <i>M</i>.<i>tb</i> induces Mcl-1 expression through PPARĪ³, which requires 15-LOX, cPLA<sub>2</sub>, and the mannose receptor (MR) [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1007100#ppat.1007100.ref004" target="_blank">4</a>]. 15-LOX and PPARĪ³, through regulation of Mcl-1, contribute to <i>M</i>.<i>tb</i>ā€™s ability to limit apoptosis and grow in human macrophages.</p

    Identification of novel PPARĪ³ effectors with NanoString.

    No full text
    <p>MDMs were transfected with PPARĪ³ (siP) or scrambled control (sc) siRNA, then infected with <i>M</i>.<i>tb</i> at MOI 5 for 6 and 24 h. <b>A)</b> Representative Western blot showing knockdown efficiency, mean knockdown efficiency was 81.7 Ā± 5.5% (N = 3). <b>B and C)</b> Total RNA was extracted and NanoString analysis was performed with a Human Immunology Panel. Shown are genes that displayed a significant (<i>p</i> < 0.05) mean fold change of at least 1.5 after PPARĪ³ knockdown and 6 <b>(B)</b> or 24 <b>(C)</b> h of infection. Asterisks in C indicate Bax and Mcl-1. N = 3. <b>D and E)</b> Total RNA was collected and gene expression analyzed by qRT-PCR. Results are expressed as Bax <b>(D)</b> and Mcl-1 <b>(E)</b> expression relative to the scrambled control cells and are the mean Ā± SEM of 3, in triplicate, * <i>p</i> < 0.05.</p

    PPARĪ³ induces Mcl-1 expression.

    No full text
    <p><b>A and B)</b> MDMs were treated with 100 nM rosiglitazone without <b>(A)</b> or with <b>(B)</b> 1h pre-treatment with GW9662. After 24 h, total RNA was collected and Mcl-1 gene expression analyzed by qRT-PCR. Results are the mean Ā± SEM of N = 5 <b>(A)</b> or 2 <b>(B)</b>. <b>C, D, F)</b> RAW cells were transfected with wild-type <b>(</b>wt; <b>C, D, F)</b> and mutated <b>(F)</b> Mcl-1 promoter luciferase reporter constructs, with and without PPARĪ³ expression plasmids. <b>C)</b> Western blot was performed to confirm PPARĪ³ expression. <b>D and F)</b> After transfection, cells were stimulated with 100 nM rosiglitazone. After 24 h, luciferase activity was determined and normalized to total protein. Results are expressed as (RLUs) normalized to cells not expressing PPARĪ³ and are the mean Ā± SEM of N = 14 <b>(D)</b> or 2ā€“4 <b>(F)</b>, * indicate a significant difference between wt reporter and the indicated condition. <b>E)</b> Schematic of Mcl-1 promoter region upstream of the luciferase gene (luc) in pGL3. The six putative PPREs are indicated, wt sequences are shown on the top with red indicating consensus to PPRE (AGGTCAnAGGTCA), with mutated sequences below and X indicating no change. <b>G)</b> RAW cells were transfected with wt or mutated Mcl-1 promoter luciferase reporter constructs. After transfection, cells were stimulated with 1 Ī¼g/ml LPS. After 24 h, luciferase activity was determined and normalized to total protein. Results are expressed as relative luminescence units (RLUs) normalized to cells not stimulated with LPS and are the mean Ā± SEM of N = 2ā€“3. <b>A-F)</b> * <i>p</i> < 0.05, ** <i>p</i> < 0.01.</p

    PPARĪ³ and Mcl-1 limit apoptosis during <i>M</i>.<i>tb</i> infection.

    No full text
    <p><b>A, B, D, E)</b> MDMs were transfected with Mcl-1 <b>(A and B),</b> PPARĪ³ <b>(D and E)</b>, or scrambled control (sc) siRNA then infected with fluorescent <i>M</i>.<i>tb</i> at MOI 50 for 24 h <b>(B),</b> or MOI 5 for 48 h <b>(D and E)</b>. Due to the variability amongst donors, these different conditions were necessary to see low levels of apoptosis in the scrambled control cells, mean 8.66 Ā± 3.69% <b>(B)</b> and 2.30 Ā± 1.69% <b>(E)</b>. <b>A)</b> Western blot showing Mcl-1 knockdown efficiency, mean knockdown efficiency was 76.6 Ā± 5.47% (N = 5). <b>B and E)</b> Data are representative of 3 experiments and are expressed as percentage of TUNEL<sup>+</sup> MDMs and are the mean Ā± SD. The cumulative increase in TUNEL<sup>+</sup> MDMs following knockdown (N = 3) is shown in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1007100#ppat.1007100.s004" target="_blank">S4A and S4B Fig</a>. <b>C)</b> MDMs were treated with 5 Ī¼M staurosporine overnight then fixed and TUNEL staining performed. <b>C and D)</b> Representative images of TUNEL staining, with TUNEL staining indicated in red and fluorescent <i>M</i>.<i>tb</i> in green. <b>F, G, H)</b> MDMs were transfected with PPARĪ³ or scrambled control (sc) siRNA <b>(F)</b> or pre-treated with GW9662 <b>(G)</b> or PD146176 <b>(H)</b> for 1 h, then infected with <i>M</i>.<i>tb</i> at MOI 5 for 24 h. MDMs were also treated with 5 Ī¼M staurosporine for 24 h. Cell death was determined with the CellTiter Glo Assay, data are expressed as % cell death, with uninfected cells set to 0%. Results are mean Ā± SEM of N = 3 <b>(F)</b> or 2 <b>(H)</b>, or representative of n = 5 <b>(G)</b>. <b>A-H)</b> * <i>p</i> < 0.05, ** <i>p</i> < 0.01, *** <i>p</i> < 0.001, **** <i>p</i> < 0.0001.</p

    Mcl-1 inhibition limits <i>M</i>.<i>tb</i> growth.

    No full text
    <p><b>A)</b> MDMs were infected with <i>M</i>.<i>tb</i> at MOI 1, then treated with the indicated Mcl-1 inhibitors. After 4 d, cells were lysed and CFU enumerated. <b>B)</b> MDMs were infected with <i>M</i>.<i>tb</i>-lux at MOI 1, then treated with the indicated Mcl-1 specific inhibitors. <i>M</i>.<i>tb</i> luciferase activity was measured over time. <b>C)</b> MDMs were infected with <i>M</i>.<i>tb</i>-lux at MOI 1, treated with 100 nM Q-VD-OPH (QVD) for 1 h, then 30 Ī¼m A-1210477 and <i>M</i>.<i>tb</i> luciferase activity was measured after 3 d. Results are the mean Ā± SD of a representative experiment of 2, in triplicate. <b>D)</b> Human PBMCs were infected with <i>M</i>.<i>tb</i> at MOI 1 to generate <i>in vitro</i> TB granulomas, and after 1 day, treated with the indicated Mcl-1 inhibitors (30 Ī¼M). After 6 d with inhibitor, cells were lysed and CFU enumerated. <b>E)</b> MDMs were infected with <i>M</i>.<i>tb</i>-lux at MOI 1, then treated with PD146176 (50 Ī¼m). After 4 d, <i>M</i>.<i>tb</i> luciferase activity was measured. <b>A-E)</b> Results are the mean Ā± SEM of N = 3 unless indicated otherwise, * <i>p</i> < 0.05, *** <i>p</i> < 0.001, **** <i>p</i> < 0.0001.</p

    <i>M</i>.<i>tb</i> induces Mcl-1 expression in human macrophages.

    No full text
    <p>MDMs <b>(A, C-F)</b> and HAMs <b>(B, G, H)</b> were infected with <i>M</i>.<i>tb</i> at MOI 5 for the indicated times <b>(A, C, D),</b> at the indicated MOI for 24 h <b>(E and F)</b>, or at MOI 5 for 24 h <b>(B, G, H)</b>. <b>A and B)</b> Total RNA was collected and gene expression of Mcl-1 analyzed by qRT-PCR. Results are expressed as Mcl-1 expression relative to uninfected cells and are the mean Ā± SEM of N = 2ā€“3. <b>C-H)</b> Protein lysates were collected and analyzed by Western blot <b>(C, E, G)</b>. Densitometry analysis of Western blots was conducted with Image J <b>(D, F, H)</b>. Data are expressed as amount of Mcl-1 protein relative to uninfected macrophages and are the mean Ā± SEM of at least 3 experiments. <b>A-H)</b> * <i>p</i> < 0.05, ** <i>p</i> < 0.01,*** <i>p</i> < 0.001, **** <i>p</i> < 0.0001.</p

    TREM1 pathway gene activation in individual HAM and MDM samples at 2 h post <i>M</i>.<i>tb</i> infection.

    No full text
    <p>The heatmap color and intensity indicate the ratio of mRNA expression <i>M</i>.<i>tb</i>/control cells. Red indicates a positive activation z-score and blue is a negative z-score. IL-6 and GM-CSF (CSF2) expression stand out in all 3 HAM samples at 2 h whereas multiple other genes are more highly differentially expressed in MDM.</p

    Linkage disequilibrium or LD heatmap of SNPs around the <i>IL-10</i> gene locus that are either recognized as eQTLs in GTEx or as GWAS hits in dbGaP (for any phenotype), or both.

    No full text
    <p>LD maps are constructed from Caucasian (CEU) and Yoruban (YRI) genomic SNP data, showing different haplotype configurations. The colored bars on top mark SNPs with eQTLs for IL-10 mRNA (blue), and dbGaP hits (red). The three marker SNPs highlighted (rs1518111, rs6686931, rs3024498) are marked by a * if recognized as an eQTL (black) or a dbGaP hit (red). Only rs1518111has both associations along with other variants in its LD block (white squares), while rs6686931and rs3024498 are only eQTLs. rs6686931 is in partial LD with rs1518111 in CEUs but in lower LD in Yorubans (YRIs). GWAS variants are part of two separate blocksā€”one separate from all eQTLs and one part of the LD block marked by rs1518111.</p
    corecore