14 research outputs found

    Phenotypic and molecular analysis of the compound <i>in cis Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> mouse model.

    No full text
    <p>(A) Schematic illustration of the chr. 18 LOH event in intestinal tumors from <i>in cis Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> mice leading to loss of both <i>Smad4</i> and <i>Apc</i> wild-type alleles. (B) Comparative phenotypic analysis of the intestinal and mammary tumor predisposition among <i>Apc</i><sup>+/1572T</sup>, <i>Smad4</i><sup>+/Sad</sup>, and <i>Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> mice. <i>Notes</i>: (1) The incidence of GI tumors was calculated after exclusion of the pyloric lesions as these present in clusters often difficult to count. (2) The multiplicity of GI tumors was calculated based on all animals with the exception of those where the high tumor burden made the count not feasible. The asterisks indicate that the apparent absence of intestinal tumor in <i>Smad4</i><sup>+/Sad</sup> control animals is not in contradiction with what previously published. These mice were sacrificed at time points matched with the ages at which compound <i>Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> mice had to be sacrificed due to the high GI and mammary tumor burden (♀: 90.4 days +/−28.4; ♂: 118.5 days +/−26.2). However, in <i>Smad4</i><sup>+/Sad</sup> animals the majority of the tumors appear at 9 months of age <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1000547#pgen.1000547-Alberici1" target="_blank">[15]</a>. (C) H&E staining of intestinal tumor sections from <i>Apc</i><sup>+/1572T</sup> (top), <i>Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> (middle), and <i>Smad4</i><sup>+/Sad</sup> (bottom) mice. (D) Smad4 IHC analysis of two intestinal adenomas from <i>Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> mice showing loss of Smad4 expression. LOH was observed in 100% of the polyps (n = 15) analyzed. PCR–based LOH analysis of the same cohort of <i>Apc</i><sup>+/1572T</sup>/<i>Smad4</i><sup>+/Sad</sup> polyps revealed loss of wild-type <i>Apc</i> allele in 87% of the cases (13/15; data not shown).</p

    Differentiation.

    No full text
    <p>HE staining of mammary tumors (A–B) and pulmonary metastases (C) from <i>Apc</i><sup>+/1572T</sup> mice shows typical mammary glandular architecture and squamous differentiation. (D–F) Luminal epithelial differentiation as shown by cytokeratin 8 (Ck8) IHC staining. (G–I) Myoepithelial differentiation revealed by IHC staining with the Sma antibody. (J–L) IHC analysis with antibodies directed against cytokeratin 14 (Ck14) confirm the presence of squamous differentiation (hair follicle and skin cellular types). (M–O) β-catenin IHC analysis shows heterogeneous subcellular localization and intracellular accumulation with fewer cells characterized by positive nuclear staining. The results shown in this figure were confirmed in 12 independent primary tumors.</p

    Biochemical characterization of the targeted <i>Apc</i>1572T allele.

    No full text
    <p>(A) Schematic representation of the APC tumor suppressor protein, its functional domains, and the truncated proteins resulting from the <i>Apc</i>1572T, <i>Apc</i>1638N, and <i>Apc</i>1638T targeted alleles. Only residual amounts (2%) of the truncated Apc1638N protein are encoded by the targeted allele, as shown by immuno-precipitation analysis of <i>Apc</i><sup>1638N/1638N</sup> ES lines <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1000547#pgen.1000547-Kielman1" target="_blank">[5]</a>. (B) β-catenin/TCF reporter assay (TOP-FLASH) analysis of <i>Apc</i><sup>+/+</sup> (1) and <i>Apc</i>-mutant ES cell lines: Apc<sup>1638T/1638T</sup> (2); Apc<sup>1572T/1572T</sup> (3); Apc<sup>1638N/1638N</sup> (4). Each bar represents the average measurement of the luciferase units from triplicate assays. For each cell line, 3 independent experiments were performed with the TOP (filled bars) and FOP (empty bars) reporter constructs. The bold figures represent the average TOP/FOP ratio of all independent experiments. Depicted error bars correspond to standard deviation. In brief, ES cells were plated on dishes coated with MEFs and subsequently transfected by lipofection with either the TOP-FLASH or FOP-FLASH reporter constructs <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1000547#pgen.1000547-Korinek1" target="_blank">[10]</a> together with the Renilla luciferase vector for normalization purposes. (C) Immuno-precipitation (IP) analysis of Apc-bound β-catenin in <i>Ap</i>c-mutant ES cell lines. For comparative purposes, immuno-precipitates obtained from equal amounts of total cellular lysates were loaded.</p

    Overview of the phenotypic comparisons between <i>Apc</i><sup>+/1572T</sup> and other <i>Apc</i>-mutant mouse models.

    No full text
    <p>Notes: Incidence is given as percentage of affected animals.</p>1,3<p>Animals were sacrificed when signs of discomfort were apparent and/or when tumor size reached 2 cm.</p>2<p>The incidence of GI tumors was calculated after exclusion of the pyloric lesions as these present in clusters often difficult to count.</p>3<p>The multiplicity of GI tumors was calculated based on all animals with the exception of those where the high tumor burden made the count not feasible.</p>*<p>This specific animal was found to carry a single tumor at 21 months of age, likely to represent a sporadic case. Background (B) of the different strains analyzed: F1: C57Bl6/J x 129Ola; Ola: inbred 129Ola; B6: inbred C57Bl6/J. n.d. not determined.</p

    Teratoma formation assays indicate an intermediate differentiation defect in <i>Apc</i><sup>1572T/1572T</sup> ES cells.

    No full text
    <p>(A) HE analysis of normal mammary gland with luminal cells surrounded by a basal layer of myoepithelial cells. (B) IF analysis of normal mammary glands for Ck8 (luminal cells, green) and Sma (myoepithelial, red). (C) HE staining of <i>Apc</i><sup>1572T/1572T</sup> teratoma showing the typical mammary gland architecture with lobular and ductal structures. (D) IF analysis of <i>Apc</i><sup>1572T/1572T</sup> teratomas for luminal and myoepithelial cell types. The frequency of these structures in teratomas derived from <i>Apc</i><sup>1572T/1572T</sup> ES cells is largely increased when compared with (F) teratomas derived from <i>Apc</i><sup>+/+</sup> ES cells. (E) Summary of the results of the teratoma differentiation assays of <i>Apc</i>-mutant ES cells. Antibodies employed to evaluate differentiation are: Glial Fibrillary Acidic Protein (GFAP) for glial cells; 2H3 for neurofilaments; SV2 for synaptic vesicles; A4.1025 for adult myosin. Mammary gland structures were primarily identified by HE and then confirmed by IF as shown in panels a–d; also in the case of cartilage and epithelial structures HE stained section were employed. n.d. not determined. Differentiation levels were scored as: (−) not present; (−*) vestigial presence; (+) present; (++) highly abundant. The shaded areas indicate groups of teratomas for which the corresponding antibody staining was negative.</p

    LOH analysis of <i>Apc</i><sup>+/1572T</sup> mammary adenocarcinomas.

    No full text
    <p>(A) PCR–based LOH analysis of tumour DNA samples amplified in the presence of radioactive nucleotides as previously described <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1000547#pgen.1000547-Smits3" target="_blank">[35]</a>. Samples were scored as having lost the wild type allele when the ratio between the intensity of the two alleles was ≤0.6. <i>Apc</i><sup>+/1572T</sup> mammary tumor samples (T); <i>Apc</i><sup>+/+</sup> DNA control (C<sub>1</sub>); <i>Apc</i><sup>+/1572T</sup> DNA control (C<sub>2</sub>). Out of the 27 samples (collected from 23 tumours) analysed, only two (lanes marked by an asterisks) show a ratio higher than 0.6 and were accordingly scored as not having allelic imbalance. (B) Western analysis of tumour-derived total protein lysates confirms the somatic loss of full length Apc. Tumour samples (T); wild type control (C<sub>3</sub>); <i>Apc</i><sup>+/1572T</sup> control from tail sample (C<sub>4</sub>).</p

    Phenotypic characterization of <i>Apc</i><sup>+/1572T</sup> mice: mammary adenocarcinomas are composed by mixed differentiation lineages with heterogeneous patterns of β-catenin intracellular accumulation and subcellular localization.

    No full text
    <p>Survival curves of (A) female and (B) male <i>Apc</i><sup>+/1572T</sup> mice, respectively. The black, green and red lines are representative of mice in the 129Ola, F1 B6x129Ola, and B6 respectively. Please note that in these graphs, age of death represents the moment at which, due to the presence of signs of discomfort or because the tumor size exceeded 2 cm<sup>3</sup>, mice had to be euthanized according to institutional and national regulations. (C) Macroscopic image of the appearance of the mammary adenocarcinomas characteristic of the <i>Apc</i>1572T model. (D) Examples of global digital microscopy scans of two mammary adenocarcinomas from <i>Apc</i><sup>+/1572T</sup> mice illustrative of the multi-lineage nature of these lesions.</p

    Transgenic expression of Bmi1 is sufficient to induce intermediate and anterior lobe pituitary tumors but does not induce medulloblastoma or glioma.

    No full text
    <p>(A) Kaplan Meier survival curves of mice carrying tumors because of GCre induced transgenic expression of Bmi1 complemented with loss of Rb. These data show that Bmi1 transgenic mice develop pituitary tumors after about one year. Pituitary tumors are also observed in Rb<sup>Lox/Lox</sup> and Bmi1<sup>LSL</sup>;Rb<sup>Lox/Lox</sup> transgenic mice. (B) Histograms showing the relative frequency and penetrance of tumors generated by the individual genotypic groups. All genotypes shown are GCre positive. Total cohort size: GCre;Bmi1<sup>LSL</sup> n = 14, GCre;Rb<sup>Lox/Lox</sup> n = 20, GCre;Rb<sup>Lox/Lox</sup>;Bmi1<sup>LSL</sup> n = 12, GCre;p53<sup>Lox/Lox</sup>;Rb<sup>Lox/Lox</sup> n = 6, GCre;p53<sup>Lox/Lox</sup>;Rb<sup>Lox/Lox</sup>; Bmi1<sup>LSL</sup> n = 4, WT mice did not develop tumors, n = 7. (C) IHC results showing transgenic Bm1 expression in tumors raised on a GCre;Bmi1<sup>LSL</sup> background (5/5), no expression in tumors raised on a Gcre;Rb<sup>lox/lox</sup> background (0/5) while some of the GCre;Bmi1<sup>LSL</sup>; Rb<sup>lox/lox</sup> mice were positive (2/5, 40%). These results are summarized in a histogram (D). Bar is 50 µm.</p
    corecore