42 research outputs found

    Nano-thin walled micro-compartments from transmembrane protein-polymer conjugates

    No full text
    The high interfacial activity of protein–polymer conjugates has inspired their use as stabilizers for Pickering emulsions, resulting in many interesting applications such as synthesis of templated micro-compartments and protocells or vehicles for drug and gene delivery. In this study we report, for the first time, the stabilization of Pickering emulsions with conjugates of a genetically modified transmembrane protein, ferric hydroxamate uptake protein component A (FhuA). The lysine residues of FhuA with open pore (FhuA ΔCVFtev) were modified to attach an initiator and consequently controlled radical polymerization (CRP) carried out via the grafting-from technique. The resulting conjugates of FhuA ΔCVFtev with poly(N-isopropylacrylamide) (PNIPAAm) and poly((2-dimethylamino)ethyl methacrylate) (PDMAEMA), the so-called building blocks based on transmembrane proteins (BBTP), have been shown to engender larger structures. The properties such as pH-responsivity, temperature-responsivity and interfacial activity of the BBTP were analyzed using UV-Vis spectrophotometry and pendant drop tensiometry. The BBTP were then utilized for the synthesis of highly stable Pickering emulsions, which could remain non-coalesced for well over a month. A new UV-crosslinkable monomer was synthesized and copolymerized with NIPAAm from the protein. The emulsion droplets, upon crosslinking of polymer chains, yielded micro-compartments. Fluorescence microscopy proved that these compartments are of micrometer scale, while cryo-scanning electron microscopy and scanning force microscopy analysis yielded a thickness in the range of 11.1 ± 0.6 to 38.0 ± 18.2 nm for the stabilizing layer of the conjugates. Such micro-compartments would prove to be beneficial in drug delivery applications, owing to the possibility of using the channel of the transmembrane protein as a gate and the smart polymer chains as trigger switches to tune the behavior of the capsules

    FXR Controls the Tumor Suppressor NDRG2 and FXR Agonists Reduce Liver Tumor Growth and Metastasis in an Orthotopic Mouse Xenograft Model

    No full text
    <div><p>The farnesoid X receptor (FXR) is expressed predominantly in tissues exposed to high levels of bile acids and controls bile acid and lipid homeostasis. FXR<sup>−/−</sup> mice develop hepatocellular carcinoma (HCC) and show an increased prevalence for intestinal malignancies, suggesting a role of FXR as a tumor suppressor in enterohepatic tissues. The N-myc downstream-regulated gene 2 (NDRG2) has been recognized as a tumor suppressor gene, which is downregulated in human hepatocellular carcinoma, colorectal carcinoma and many other malignancies.</p> <p>We show reduced NDRG2 mRNA in livers of FXR<sup>−/−</sup> mice compared to wild type mice and both, FXR and NDRG2 mRNAs, are reduced in human HCC compared to normal liver. Gene reporter assays and Chromatin Immunoprecipitation data support that FXR directly controls NDRG2 transcription via IR1-type element(s) identified in the first introns of the human, mouse and rat NDRG2 genes. NDRG2 mRNA was induced by non-steroidal FXR agonists in livers of mice and the magnitude of induction of NDRG2 mRNA in three different human hepatoma cell lines was increased when ectopically expressing human FXR. Growth and metastasis of SK-Hep-1 cells was strongly reduced by non-steroidal FXR agonists in an orthotopic liver xenograft tumor model. Ectopic expression of FXR in SK-Hep1 cells reduced tumor growth and metastasis potential of corresponding cells and increased the anti-tumor efficacy of FXR agonists, which may be partly mediated via increased NDRG2 expression. FXR agonists may show a potential in the prevention and/or treatment of human hepatocellular carcinoma, a devastating malignancy with increasing prevalence and limited therapeutic options.</p> </div

    Induction of NDRG2 in human hepatoma cell lines.

    No full text
    <p><b>A</b>, Ectopic expression of FXR in HepG2-FXR5 compared to HepG2 parental cells. Positions of bands representing FXR and GAPDH proteins in the western blots from aliquots of whole cell extracts are indicated. <b>B</b>, Dose dependent induction of NDRG2 mRNA in HepG2 and HepG2-FXR5 cells by the FXR agonist PX20350. HepG2 or HepG2-FXR5 cells were grown for 18 h in medium containing either DMSO, 0.5 µM of PX20350 or 0.5 µM of PX20606 in 96 well plates as indicated and relative fold induction of NDRG2 mRNA over the DMSO control was determined by RT-qPCR using the ΔΔCt method. <b>C</b>, Induction of NDRG2 protein by the FXR agonists Px20350 and PX20606 in HepG2-FXR5 cells. HepG2-FXR5 cells were grown to a density of around 60% and further cultivated for 18 h in medium supplemented with either DMSO as a vehicle, 0.5 µM PX20350 or 0.5 µM PX20606 as indicated and aliquots of whole cell extracts analysed by western blots. Relative positions of size marker protein bands and the position of NDRG2 and GAPDH proteins are indicated. <b>D</b>, Stable overproduction of FXR in HuH-7-37 compared to HuH-7 cells. Positions of bands representing FXR and GAPDH proteins analysed by western blots from aliquots of whole cell extracts are indicated. <b>E</b>, Dose dependent induction of NDRG2 mRNA in HuH7 and HuH7-37 cells by FXR agonist Px20350. HuH7 or HuH7-37 cells were grown for 18 h in medium containing either DMSO or increasing concentrations of PX20350 in DMSO in 96 well plates and relative fold induction over DMSO control of NDRG2 mRNA was determined by RT- qPCR using the ΔΔCt method. <b>F</b>, NDRG2 is a direct transcriptional target of FXR. HepG2-FXR5 cells, grown in quadruplicates in 12 well plates at a density of around 60%, were further incubated for 4 hours in growth medium supplemented with either DMSO, 0.5 µM PX20350 (PX) or 0.5 µM PX20350 together with 10 µM Cycloheximide (CHX). The relative fold induction of NDRG2- and SHP- mRNAs over DMSO as control were determined by RT- qPCR using the ΔΔCt method and a Student's t-test was performed (*** = p<0.001).</p

    Identification of functional IR1 site(s) in NDRG2.

    No full text
    <p><b>A</b>, Schematic representation of the NDRG2 gene with the IR1 site(s) located in the first intron. Human, mouse and rat intronic sequences with IR1 elements (in bold) are shown below (IR1 elements start at positions +1560, +757 and +1394 respectively). A version of the human NDRG2 response element with mutated IR1 sites is shown below. <b>B</b>, Luciferase reporter assays from HEK 293 cells co-transfected with pTReX-Dest30-hFXRα2 and a luciferase reporter plasmid containing either one copy of the wild type IR1 element (IR1) or the mutated element (IR1<sup>mut</sup>) from the human NDRG2 gene or 2 copies of the human IBABP-RE (IBABP). The fold induction in presence of 0.5 µM Px20350 (Px) versus DMSO control (D) of firefly luciferase activities (normalized against renilla luciferase) of triplicate experiments is shown. <b>C</b>, Amplification of a 196 bp DNA fragment encompassing the IR1 sequences from chromatin prepared by ChIP from HepG2 or HepG2-FLAG-FXR cells. PCR reactions from input DNA and DNA derived from the immunoprecipitation with the M2 anti-FLAG antibody are indicated <b>D</b>, Localization of murine FXRα and RXRα by ChIP-seq to a site corresponding to the predicted IR1 recognition element (star) in the first intron of the mouse Ndrg2 gene on chromosome 14 in mouse liver. The FXRα and RXRα UCSC genome browser tracks shown for the NDRG2 gene were retrieved from genome wide ChIP-seq datasets published by Thomas et al. <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0043044#pone.0043044-Thomas1" target="_blank">[22]</a> and Boergesen et al. <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0043044#pone.0043044-Boergesen1" target="_blank">[41]</a>, respectively. <b>E</b>, Chemical structures of FXR agonists. Among different structural changes, the unfavourable stilbene linker present in GW4064 was replaced by an amino-methylene linker in Px20350 <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0043044#pone.0043044-Abel1" target="_blank">[40]</a>. The further modified Px20606 <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0043044#pone.0043044-Kremoser1" target="_blank">[53]</a> yielded improved <i>in vivo</i> pharmakokinetic properties compared to Px20350. A characterization of FXR agonists in <i>in-vitro</i> assays is presented in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0043044#pone-0043044-t001" target="_blank">Table 1</a>.</p
    corecore