11 research outputs found

    Deregulated LMα5 expression in the intestine of patients with tufting enteropathy and collagenous colitis.

    No full text
    <p>LMα5 detection on small intestine (A) and colon (B) from patients with tufting enteropathy and collagenous colitis reveal an abnormal location of this chain as compared to controls. While α5 is detected mostly at the villus compartment in control specimen, it is found also in the crypt region (inset) in tufting enteropathy specimen. In a specimen of collagenous colitis, the staining is stronger all over the crypt-villus axis. Arrows point to the BM region.</p

    Presence of laminin-511 inhibits TOPflash activity.

    No full text
    <p>(A) <i>In situ</i> hybridization of Msx1 on embryonic control and KO intestines showing that Msx1 is stimulated in knockout endodermal cells (arrow); e: endoderm; m: mesenchyme. (B) Gene expression ratios determined by RT-qPCR of Pitx2 and Sfrp2 between intestinal E15.5 knockout and control tissues, and on isolated mesenchymal or endodermal compartments confirm the increase of both molecules in the absence of laminin α5; for further details see legend to <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0037710#pone-0037710-g002" target="_blank">figure 2</a> (mean +/− SEM, n = 7–9; * p<0.02). (C) HEK293 cells and lentiviral <i>lama5</i> shRNA m-IC<sub>Cl2</sub> infected cells seeded on plastic, laminin-111, cell-derived laminin-511 (LM-511C) or on recombinant laminin-511(LM-511R) were transfected with TOPflash or the negative FOPflash vector. The graphs represent the average relative luciferase activity normalized to luciferase Renilla activity; this ratio was then normalized to that obtained on plastic (n = 5, n = 3 for HEK293 on laminin-111, n = 1 for laminin-511(R); in duplicate; mean +/− SEM). For each cell line, TOPflash activity on laminin-111 does not statistically differ to that observed on plastic. Note that the TOPflash activity is statistically inhibited when cells are grown on laminin-511 as compared to laminin-111 (* p<0.05; *** p<0.001).</p

    Cell survival and activation of Akt in epithelial cells adhering to laminin-511.

    No full text
    <p>(A) m-IC<sub>Cl2</sub> epithelial cells and (B) embryonic smooth muscle cells (SMC) were plated on uncoated dishes +/− EGF and insulin, and on dishes containing laminin-511 (LM-511) or laminin-111 (LM-111). The PI3K inhibitor wortmannin was added where indicated. Cell lysates were analyzed by western blotting for phosphorylated Akt (P-Akt), Akt (pan-Akt) and actin. Note that activation of Akt is detectable in epithelial cells cultured on laminin-511 but not on laminin-111 (see quantification of the representative gel). No activation occurred in smooth muscle cells in the presence of laminin-511. In parallel, epithelial (A) and smooth muscle cells (B) were photographed by phase contrast microscopy on laminin-511 matrix (LM-511) and on laminin-111 (LM-111) with or without wortmannin (Wm). Cell spreading on laminin-511 (left pictures) versus laminin-111 (right pictures) was confirmed by flattening of the cells and reorganization of the cytoskeleton as probed with TRITC-phalloidin to visualize F-actin. (C) Representative immunoblots showing the expression of Akt (pan-Akt), Akt2 and Phospho-Akt (P-Akt) in E15.5 control (WT) and knockout (KO) intestines and quantification of two independent experiments as ratio between KO (grey bars) and WT (black bars) intestines. Data were normalized using actin.</p

    Laminin-511 controls survival and epithelial cell migration.

    No full text
    <p>(A) In a survival assay, m-IC<sub>Cl2</sub> intestinal cells were cultured with H<sub>2</sub>O<sub>2</sub> on laminin-111 (LM-111), laminin-511 coated-dishes or on laminin-511 (LM-511) with wortmannin (Wm). Survival rates, as ratios normalized to plastic, were determined by a MTS assay. Note a better cell survival rate on laminin-511 as compared to laminin-111, which is abolished upon treatment with Wm (mean +/− SEM, n = 5) (*** p<0.001). Immunofluorescence pictures (right) show more caspase-3-positive cells (arrows) on uncoated dishes (control) as compared to laminin-511. (B) Both migration velocity and cumulative migration distance of cells are significantly enhanced when m-IC<sub>Cl2</sub> cells are seeded on laminin-511 (LM-511; +/− wortmannin: Wm) versus laminin-111 (LM-111) or uncoated dishes (control) (mean +/− SEM, n = 5) (*** p<0.001). (C) Chemotactic migration of m-IC<sub>Cl2</sub> cells was visualized by phase contrast microscopy and cell counting on uncoated dishes (control), laminin-111 (LM-111), and laminin-511 (LM-511) in the presence or absence of wortmannin (Wm). Note that laminin-511 stimulated significantly cell migration independently of the PI3K/Akt pathway. In both assays, wortmannin did not affect laminin-511 enhanced migration. The dotted line represents the starting point of migration. Data (n≥5) are given as mean +/− SEM;** p<0.01; *** p<0.001.</p

    Schematic of the role of laminin-511 in intestinal tissue homeostasis.

    No full text
    <p>Laminin-511 is deposited in the intestine by both epithelial and mesenchymal cells <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0037710#pone.0037710-Lefebvre1" target="_blank">[11]</a>. Its absence (KO model or RNA knockdown experiments) or presence (<i>in vitro</i> assays) leads to activation or inactivation of Wnt and PI3Kinase signaling pathways promoting multiple cellular responses that impact on survival, migration and differentiation.</p

    Muscle differentiation genes are regulated by laminin α5 chain.

    No full text
    <p>(A) Semi-quantitative RT-PCR experiments were performed on E15.5 control and knockout intestines for genes belonging to the muscle compartment. Data are presented as fold changes between knockout (grey bars) and wild-type (black bars) (mean +/− SEM, n = 5 to n = 9) (* p<0.05). (B) Immunostaining of MyoD1 on E15.5 control and KO intestines or on derived-cultured mesenchymal cells shows that MyoD1 is induced in knockout mesenchymal cells (arrows). Nuclei are stained with DAPI (blue). (C) Expression of Hlx1 by RT-qPCR showing that its expression is enhanced in <i>lama5</i> deficient versus wild-type intestines (mean +/− SEM, n = 6) (* p<0.05). Quantitative RT-PCR was performed on separated endodermal and mesenchymal compartments. The diagram shows the relative expression of Hlx1 between mesenchyme (mes) and endoderm (endo) with value 1 representing the total amount in wild-type intestines. Expression of Hlx1 is increased specifically in the mesenchymal compartment of LMα5<sup>−/−</sup> intestines. (D) Effect of <i>lama5</i> siRNA on mesenchyme-derived target gene expression: Hlx1 (a, c) and MyoD1 (b, d). Embryonic mesenchymal cells (panels a and b) and adult intestinal smooth muscle cells (panels c and d) were cultured in the presence of control- and <i>lama5</i>-siRNA, respectively. <i>Lama5</i>-siRNA decreases LMα5 gene (up to 68%) and protein expression (in green) in both embryonic and adult cells. Note that <i>lama5</i>-siRNA upregulates Hlx1 gene expression and MyoD protein expression. After 72 h, gene expression was analyzed by RT-qPCR upon normalization to GAPDH and is expressed as relative fold-change (mean +/− SEM; n = 3) compared to control-siRNA. Arrows point at MyoD positive cells. Nuclei are stained with DAPI.</p

    Important role of p53 in regulating LM expression in IBD and colitis-associated cancer.

    No full text
    <p>We propose the following scenario where inflammation triggers nuclear accumulation of p53 that transactivates expression of LMα1 and its deposition to the BM concomitantly to that of LMα5. Overexpression of LMα5 in response to inflammation is a p53 independent mechanism. In a chronically inflamed microenvironment highly expressed LMα1 and LMα5 may provide physical barrier function resulting in attenuated inflammation as demonstrated in transgenic mice. Yet, high LMα1 and LMα5 in a carcinogenic setting subsequent to chronic colitis may contribute to a pro-tumorigenic microenvironment.</p

    Laminins protect from inflammation.

    No full text
    <p>(<b>A–B</b>) Histological views of colon tissue (stained with PAS) and expression of LMα5 and LMα1 on cryosections of colon from parental, Tg-<i>lama5</i> or Tg-<i>lama1</i> mice untreated or treated with DSS. This showed that both chains were ectopically expressed in the glandular crypt region in the transgenic animals (arrows) and that LMα1 expression was further extended by DSS treatment. (<b>C</b>) Inflammatory scores (mean +/− SEM; n = 5) assessed on the Swiss-roll of the colon and rectum of transgenic <i>lama5</i> and <i>lama1</i> animals (black columns) as compared to controls (grey columns), all treated with DSS. (<b>D</b>) Levels of pro-inflammatory cytokines (mean +/− SEM; n = 6) measured by ELISA in protein extracts from distal colon of DSS-treated parental (grey columns) and DSS-Tg-<i>lama1</i> or DSS-Tg-<i>lama5</i> (black columns) mice. Data were normalized to the mean of parental values as levels of cytokines turned out to be mouse strain-dependent. Statistical differences were compared to parental mice. e: epithelial cells; lp: <i>lamina propria</i>; mm: <i>muscularis mucosae</i>. Nuclei are visualized with DAPI. *<i>p</i><0.05, **<i>p</i><0.01. Scale bars: 50 µm.</p

    Colitis-associated tumor development is increased in transgenic mice overexpressing LMα1.

    No full text
    <p>(<b>A</b>) Schematic overview of the AOM/DSS and of the cyclic DSS protocols. (<b>B</b>) Control and transgenic mice treated with AOM/DSS or with cyclic DSS develop different types of lesions among them dysplasia and <i>in situ</i> carcinomas that were quantified. Tg-<i>lama1</i> animals develop more tumors than controls when submitted to the treatments (n = 7, *p<0.05 for AOM/DSS; n = 7, *p<0.05 for cyclic DSS). (<b>C</b>) Dysplasia and an <i>in situ</i> carcinoma are shown by hematoxylin-eosin staining (HE). Glands in these lesions are strongly positive for LMα1 (red), LMα5 (green, lower panels) and present nuclear p53 (green, middle panels). Nuclei were visualized with DAPI. e: epithelial cells; m: muscle; c: cancer cells; s: stroma; arrows: BM area. Scale bars: 50 µm.</p

    Inflammation response triggers expression of LMα1/α5 and nuclear p53 accumulation in human and murine colitis.

    No full text
    <p>(<b>A</b>) Expression of the LMα5 and LMα1 chains in normal mucosa (control), in mild inflamed glands and around UACL from colon of patients with IBD. Immunostaining for LMα5 in IBD samples is extended along the colonic glands with a strong staining at the deeper crypt region and is highly expressed in UACL (arrows). LMα1 is detected selectively at the BM at the crypt bottom in IBD samples and in UACL while it is absent from uninflamed regions; unspecific cytoplasmic immunoreactivity is seen in goblet cells (g). Inset: higher magnification of the deeper crypt region. (<b>B</b>) Expression of LMα5 and LMα1 in cryosections of colon from control and DSS-treated mice. Note, as in human IBD, high expression of LMα5 and LMα1 was observed in mouse colitis at the bottom of the colonic glands. (<b>C</b>) Nuclei of epithelial cells from inflamed colonic segments and in UACL of IBD patients were positive for p53 while nuclei of the adjacent normal crypts showed rare p53-positive cells scattered within the glands. (<b>D</b>) Expression of p53 (green) and LMα1 (red) in colon samples from DSS-treated mice. At day 3 after treatment, p53 immunoreactivity was present in numerous nuclei within epithelial cells while LMα1 co-staining was weak; inset: enlarged area with nuclear p53 expression. At day 5, intense LMα1 staining was observed surrounding weaker p53-positive glands (inset). (<b>E</b>) The diagram summarizes the distribution of the main LM chains found in normal colonic mucosa (control), in mild inflamed glands and around UACL from colon of patients with IBD. Note that only the staining at the epithelial BM is schematically represented for clarity. e: epithelial cells; lp: <i>lamina propria</i>; mm: <i>muscularis mucosae</i>; arrows: ectopic staining at crypt bottom and staining around UACL. Scale bars: 50 µm (human), 25 µm (mouse).</p
    corecore