27 research outputs found

    TIG3 Tumor Suppressor-Dependent Organelle Redistribution and Apoptosis in Skin Cancer Cells

    Get PDF
    TIG3 is a tumor suppressor protein that limits keratinocyte survival during normal differentiation. It is also important in cancer, as TIG3 level is reduced in tumors and in skin cancer cell lines, suggesting that loss of expression may be required for cancer cell survival. An important goal is identifying how TIG3 limits cell survival. In the present study we show that TIG3 expression in epidermal squamous cell carcinoma SCC-13 cells reduces cell proliferation and promotes morphological and biochemical apoptosis. To identify the mechanism that drives these changes, we demonstrate that TIG3 localizes near the centrosome and that pericentrosomal accumulation of TIG3 alters microtubule and microfilament organization and organelle distribution. Organelle accumulation at the centrosome is a hallmark of apoptosis and we demonstrate that TIG3 promotes pericentrosomal organelle accumulation. These changes are associated with reduced cyclin D1, cyclin E and cyclin A, and increased p21 level. In addition, Bax level is increased and Bcl-XL level is reduced, and cleavage of procaspase 3, procaspase 9 and PARP is enhanced. We propose that pericentrosomal localization of TIG3 is a key event that results in microtubule and microfilament redistribution and pericentrosomal organelle clustering and that leads to cancer cell apoptosis

    Synthesis and biochemical characterization of EGF receptor in a water-soluble membrane model system

    No full text
    <div><p>ErbB (Erythroblastic Leukemia Viral Oncogene Homolog) receptor tyrosine kinases are critical for tissue development and maintenance, and frequently become oncogenic when mutated or overexpressed. <i>In vitro</i> analysis of ErbB receptor kinases can be difficult because of their large size and poor water solubility. Here we report improved production and assembly of the correctly folded full-length EGF receptor (EGFR) into nanolipoprotein particles (NLPs). NLPs are ~10 nm in diameter discoidal cell membrane mimics composed of apolipoproteins surrounding a lipid bilayer. NLPs containing EGFR were synthesized via incubation of baculovirus-produced recombinant EGFR with apolipoprotein and phosphoplipids under conditions that favor self-assembly. The resulting EGFR-NLPs were the correct size, formed dimers and multimers, had intrinsic autophosphorylation activity, and retained the ability to interact with EGFR-targeted ligands and inhibitors consistent with previously-published <i>in vitro</i> binding affinities. We anticipate rapid adoption of EGFR-NLPs for structural studies of full-length receptors and drug screening, as well as for the <i>in vitro</i> characterization of ErbB heterodimers and disease-relevant mutants.</p></div

    Diagnostic Microdosing Approach to Study Gemcitabine Resistance

    No full text
    Gemcitabine metabolites cause the termination of DNA replication and induction of apoptosis. We determined whether subtherapeutic β€œmicrodoses” of gemcitabine are incorporated into DNA at levels that correlate to drug cytotoxicity. A pair of nearly isogenic bladder cancer cell lines differing in resistance to several chemotherapy drugs were treated with various concentrations of <sup>14</sup>C-labeled gemcitabine for 4–24 h. Drug incorporation into DNA was determined by accelerator mass spectrometry. A mechanistic analysis determined that RRM2, a DNA synthesis protein and a known resistance factor, substantially mediated gemcitabine toxicity. These results support gemcitabine levels in DNA as a potential biomarker of drug cytotoxicity

    NLP assemblies contain EGFR monomers, dimers, and multimers.

    No full text
    <p>Empty and EGFR-NLPs were separated by SDS-PAGE with (+Ξ²-ME/heat) and without denaturing conditions (-Ξ²-ME/heat) to observe EGFR multimers and analyzed by immunoblotting with anti-pY4G10, anti-EGFR, and anti-ApoA1 antibodies. Similar results were observed in each of three biological replicates. Numbers indicate band intensity relative to EGFR-NLPs normalized to ApoA1 signal.</p

    NLP-associated EGFR interacts with EGFR-targeted therapeutic agents.

    No full text
    <p><i>A</i>, NLP incorporated EGFR was dephosphorylated via phosphatase treatment (CIP) then allowed to autophosphorylate in the absence or presence of indicated tyrosine kinase inhibitors (M, mubritinib; A, afatinib; C, canertinib; L, lapatinib) followed by immunoblotting with anti-pY4G10 and anti-EGFR antibodies. Similar results were observed in each of three biological replicates. *Autophosphorylation is observed without additional EGF due to EGF being present in the fetal bovine serum and in the initial assembly mixture. <i>B</i>, Association and dissociation of 100 nM of empty or EGFR-NLP to biotinylated EGF on the sensors with and without 1 ΞΌM unlabeled EGF or cetuximab. Data points indicate the mean of two technical duplicates and are representative of results from two biological replicates. <i>C</i>, Association and dissociation curves of 100 nM EGFR-NLPs binding to biotinylated EGF on the sensors in the presence of increasing levels of Cetuximab. Cetuximab concentrations were titrated by a 1.5-fold dilution to the concentrations displayed to the right of the graph. Curves were normalized by subtracting the 0 ΞΌM Cetuximab curve. <i>D</i>, Log plot of cetuximab inhibiting the binding of EGFR-NLPs to biotinylated EGF on the sensors. Data points indicate the mean of two technical duplicates and are representative of results from two biological replicates.</p
    corecore