15 research outputs found

    Sonication Assisted Hierarchical Decoration of Ag-NP on Zinc Oxide Nanoflower Impregnated Eggshell Membrane: Evaluation of Antibacterial Activity and in Vitro Cytocompatibility

    No full text
    Metal/metal oxide nanoparticles have long been used as an antibacterial substitute, but fabrication of an effective carrier or delivery matrix for achieving a sustain release profile with high bactericidal efficacy alongwith good cytocompatibility is still an unresolved challenge. Herein, the study demonstrates a facile and unique route to fabricate a hierarchical nanobiocomposite with effective loading of ZnO/silver nanoparticles (Ag-NPs) in order to attain excellent bactericidal efficacy with good and sustainable release profile. Surface functionalized eggshell membranes (ESM) were deployed as three-dimensional loading matrices for efficient loading of ZnO/Ag-NPs. A simple sonochemical guided approach was adopted to synthesize ZnO nanoflakes in situ onto the microfibrous ESM and decorate it with Ag-NPs to fabricate a nanobiocomposite. Microstructural analysis confirms successful anchorage of ZnO nanoflakes and Ag-NPs on microfibrous eggshell membrane thus reinstating hierarchical morphology of the nanobiocomposites. FT-IR spectra confirms the biochemical composition whereas XPS analysis ratifies the interaction between ZnO and Ag-NPs further substantiating metallic state of Ag. ICP-MS studies affirms excellent and sustainable release profile of nanoparticles from the nanobiocomposites. Owing to the synergistic activity of ZnO/Ag-NPs, the nanobiocomposites demonstrated exceptional bactericidal activity against Gram-negative, E. coli or P. aeruginosa, and Gram-positive, S. aureus or B. subtilis, bacterial cells. Moreover, inherent antibacterial property of microfibrous natural ESM contributes positively toward the overall bactericidal activity. Further, a direct exposure of nanobiocomposites with NIH 3T3 cells revealed the biocompatible nature of developed matrices. Prolonged exposure also indicated that the 3T3 cells tend to adhere onto the microfibrous nanobiocomposite without any observable deformation in cellular morphology. The architectural tribology and excellent bactericidal performance of the nanobiocomposites along with its cytocompatible nature manifests its application as an alternate platform for varying biomedical applications

    Capsaicin-induced activation of p53-SMAR1 auto-regulatory loop down-regulates VEGF in non-small cell lung cancer to restrain angiogenesis.

    No full text
    Lung cancer is the leading cause of cancer-related deaths worldwide. Despite decades of research, the treatment options for lung cancer patients remain inadequate, either to offer a cure or even a substantial survival advantage owing to its intrinsic resistance to chemotherapy. Our results propose the effectiveness of capsaicin in down-regulating VEGF expression in non-small cell lung carcinoma (NSCLC) cells in hypoxic environment. Capsaicin-treatment re-activated p53-SMAR1 positive feed-back loop in these cells to persuade p53-mediated HIF-1α degradation and SMAR1-induced repression of Cox-2 expression that restrained HIF-1α nuclear localization. Such signal-modulations consequently down regulated VEGF expression to thwart endothelial cell migration and network formation, pre-requisites of angiogenesis in tumor micro-environment. The above results advocate the candidature of capsaicin in exclusively targeting angiogenesis by down-regulating VEGF in tumor cells to achieve more efficient and cogent therapy of resistant NSCLC

    Additional file 1: Figure S1. of Aspirin inhibits epithelial-to-mesenchymal transition and migration of oncogenic K-ras-expressing non-small cell lung carcinoma cells by down-regulating E-cadherin repressor Slug

    No full text
    p53 mutation exerts increased migratory effect in combination with oncogenic K-ras-expressing system on NSCLC cells migration. Phase contrast images (left panels) depicting migration of NCI-H522 cells (oncogenic K-ras/mutant p53), mutant p53-reconstituted H1299 cells (wild type K-ras/mutated p53), A549 cells (oncogenic K-ras/wild type p53), and wild type p53 reconstituted H1299 cells (wild type K-ras/wild type p53). Graphical representation of percent cell migration in wound healing assay (right panel) with inset showing immunoblot analysis for the transfection efficiency of p53 - R175H clone in H1299 cells and p53 - cDNA in H1299 cells. Scale bar: 100 μm. (TIFF 2457 kb

    Republished: Sulphur alters NFκB-p300 cross-talk in favour of p53-p300 to induce apoptosis in non-small cell lung carcinoma

    No full text
    Adverse side effects of chemotherapy during cancer treatment have shifted considerable focus towards therapies that are not only targeted but are also devoid of toxic side effects. We evaluated the antitumorigenic activity of sulphur, and delineated the molecular mechanisms underlying sulphurinduced apoptosis in non-small cell lung carcinoma (NSCLC) cells. A search for the underlying mechanism revealed that the choice between the two cellular processes, NFκBp65-mediated survival and p53-mediated apoptosis, was decided by the competition for a limited pool of transcriptional coactivator protein p300 in NSCLC cells. In contrast, sulphur inhibited otherwise upregulated survival signaling in NSCLC cells by perturbing the nuclear translocation of p65NFκB, its association with p300 histone acetylase, and subsequent transcription of Bcl-2. Under such anti-survival condition, induction of p53-p300 cross-talk enhanced the transcriptional activity of p53 and intrinsic mitochondrial death cascade. Overall, the findings of this preclinical study clearly delineated the molecular mechanism underlying the apoptogenic effect of the non-toxic homeopathic remedy, sulphur, in NSCLC cells

    Capsaicin suppresses VEGF expression by SMAR1-mediated down regulation of Cox-2.

    No full text
    <p>(A) Control-/SMAR1-shRNA (<i>left panel</i>) or control-vector/SMAR1-cDNA (<i>right panel</i>) transfected Hy-A549 cells were treated with capsaicin were assayed for Cox-2 expression by Western blot and RT-PCR. Transfection efficiency was verified by Western blot (<i>bottom panels</i>). (B) Cox-2 promoter activity was checked in Hy-A549. Schematic representation of the Cox-2 promoter showing the probable SMAR1-binding sites predicted by using the MARWIZ software, PCR run with eight different sets of primers designed for each probable MAR binding site spreading over four regions (−141 bp to −421 bp; −631 bp to −1121 bp; −1262 bp to −1331 bp; −1471 bp to −1891 bp). ChIP assay with anti-SMAR1 was performed on MAR-binding regions of Cox-2 promoter. Input and control IgG was used as internal control and negative control. (C) The relative abundance of SMAR1 on Cox-2 promoter was analyzed in control and capsaicin treated Hy-A549 cells at binding sites 6 & 7 after ChIP of Cox-2 promoter with anti-SMAR1(<i>left panel</i>) and represented graphically (<i>right panel</i>). (D) Control-/SMAR1-shRNA-transfected Hy-A549 cells were treated with capsaicin and analyzed for reporter HIF-1α and VEGF gene expression by RT-PCR. α-Actin/GAPDH were used as internal loading control. Values are mean ±SEM of three independent experiments in each case or representative of typical experiment.</p

    Effect of capsaicin on lung cancer cell spent medium-induced endothelial cell migration and network formation.

    No full text
    <p>(A) Migration of HUVECs in presence or absence of spent media of NME, WI-38, A549 and Hy-A549 (CoCl<sub>2</sub>-stimulated to mimic hypoxic condition required for tumor-induced angiogenesis) were subjected to bi-directional wound healing assay for 24 h (<i>left panel</i>). The number of cells migrated in the wound area are represented graphically (<i>right panel</i>). (B) Representative images of HUVEC migration upon incubation with capsaicin-treated (0–50 µM) Hy-A549 cell spent medium (<i>left panel</i>). Percent cell migrated in the wound area has been represented graphically (<i>right panel</i>). (C) Hy-A549 cells were treated with capsaicin in a dose-dependent manner for 24 h and cell viability was scored by trypan blue dye-exclusion assay (<i>left panel</i>). Hy-A549 cells, treated with capsaicin (37.5 µM), were subjected to Annexin-V-FITC/PI binding and analyzed flow cytometrically for the determination of percent early apoptosis (<i>right panel</i>). (D) Cytotoxic effect of different doses of capsaisin on HUVEC cells were measured by trypan blue dye-exclusion assay. (E) Graphical representation of HUVEC migration upon incubation with spent media from capsaicin-treated (37.5 µM) HBL-100, HCT-15, HeLa and A549. (F) Representative images of capillary-like sprout formation by HUVECs in presence of media alone or spent media of WI-38/A549/Hy-A549/capsaicin-treated Hy-A549. Values are mean ± SEM of three independent experiments in each case or representative of typical experiment.</p

    Capsaicin inhibits the nuclear localization of HIF-1α by down regulating Cox-2 in a p53-dependent manner.

    No full text
    <p>(A) Immunoblot representing nuclear and cytosolic levels of HIF-1α in capsaicin-treated/p53-shRNA-transfected Hy-A549 cells. (B) HIF-1α expression was monitored in capsaicin-treated Hy-A549 cells by confocal microscopy (magnification 60x). (C) Time-dependent expression of Cox-2-mRNA and -protein in capsaicin-treated Hy-A549 cells was determined by Western blot and RT-PCR. (D) Control siRNA-/Cox-2-siRNA-transfected Hy-A549 cells were treated with capsaicin (37.5 µM; 24 h) and nuclear translocation of HIF-1α was assessed by Western blot analysis (<i>left panel</i>). Transfection efficiency was determined by analyzing the expression of Cox-2 (<i>right panel</i>). (E) Control vector-/Cox-2 cDNA transfected Hy-A549 cells were treated with capsaicin (37.5 µM; 24 h) and analyzed for nuclear and cytosolic expression of HIF-1α (<i>left panel</i>). Transfection efficiency of Cox-2 was also verified (<i>right panel</i>). (F) Control-shRNA-/p53-shRNA-transfected Hy-A549 cells were treated with capsaicin (37.5 µM; 24 h) and expression profiles of Cox-2 both at protein and mRNA level were examined. (G) Time-dependent expression profiles of SMAR1-protein and mRNA in capsaicin-treated Hy-A549 cells were determined by Western blot and RT-PCR (<i>left panels</i>). Control-/p53-shRNA transfected Hy-A549 cells were treated with capsaicin and expression levels of SMAR1 were checked (<i>right panel</i>). (H) Control-/SMAR1-shRNA transfected Hy-A549 cells were treated with capsaicin and immune-blotted with p-Ser<sup>15</sup>-p53. α-Actin/H1-Histone/GAPDH were used as internal loading control. Values are mean ± SEM of three independent experiments in each case or representative of typical experiment.</p
    corecore