16 research outputs found

    Ubiquitously excess wild-type Met in developing embryos results into hyperflexed forelimbs.

    No full text
    <p>(A) Top: <i>Del-R26</i><sup><i>Met</i></sup> and control P0 mice showing hyperflexed limbs in mutants. Bottom: anti-myosin heavy chain II IHC using MF20 antibodies on forelimb transversal sections of P0 <i>Del-R26</i><sup><i>Met</i></sup> and control mice at the level of the forearm showing almost absence of extensor (asterisk) and a great reduction of flexor (arrowhead) muscle mass in mutants. (B, C) Whole mount ISH with <i>MyoD</i> probe of E12.5 embryos (B) and β-galactosidase staining of E11.5 embryos (C) showing that developing appendicular muscles are reduced in <i>Del-R26</i><sup><i>Met</i></sup> embryos (limbs are outlined in panels). The arrowhead in bottom panel B indicates developing ventral limb muscles (flexor). Scale: 500μm.</p

    Schematic representation summarizing the different molecular and phenotypic effects of enhanced Met expression in myoblasts and limb mesenchymal cells.

    No full text
    <p>In a wild-type context (top), limb mesenchymal cells secrete HGF required for migration of myoblasts towards the limb buds. Enhanced expression of Met in myoblasts (as assessed in <i>Pax3-R26</i><sup><i>Met</i></sup> embryos; middle) does not alter their migration due to a buffering event: the activation levels of signalling effectors such as ERKs and Akt are restrained despite enhanced Met phosphorylation. The size of each signal is representative of their phosphorylation levels. Limb mesenchymal cells are vulnerable to ectopic Met expression (as assessed in <i>Prx1-R26</i><sup><i>Met</i></sup> embryos; bottom), illustrated by changes in gene expression, by failure of HGF bioavailability, and by myoblast migration defects. Alteration of HGF bioavailability can be due to: 1) upregulation of a negative interactor that would interfere with the capacity of HGF to bind/activate Met (indicated as “HGF inhibitor”), 2) downregulation of a HGF interactor acting as enhancer of its bioactivity (indicated as “HGF activator”), 3) expression of a chemorepellent factor that renders limb mesenchyme inaccessible to migrating myoblasts, or 4) HGF titration by ectopic Met in mesenchymal cells (indicated as “HGF trapping”).</p

    Ectopic Met in limb mesenchyme down-regulates the expression levels of <i>Notum</i> and <i>Syndecan4</i>.

    No full text
    <p>qRT-PCR analysis of transcript levels of mouse <i>Met</i> (<i>mMet</i>), <i>Pax3</i>, <i>Notum</i>, and <i>Syndecan4</i> (<i>Sdc4</i>) in controls (ctrl; n = 11), <i>Del-R26</i><sup><i>Met</i></sup> (Del-Met; n = 11), <i>Met</i><sup><i>LacZ/d(neo)</i></sup> (KO; n = 7). Each dots corresponds to transcript levels in forelimbs of E10.5 individual embryos (done in triplicate). Columns correspond to the average value, expressed as mean ± s.e.m. Note: downregulation of <i>mMet</i> and <i>Pax3</i> in <i>Del-R26</i><sup><i>Met</i></sup> and <i>Met</i><sup><i>LacZ/d(neo)</i></sup> mutants compared to control, consistent with lack of migrating myoblasts; downregulation of <i>Notum</i> and <i>Syndecan4</i> in <i>Del-R26</i><sup><i>Met</i></sup> mutants compared to control, whereas no significant changes were found in <i>Met</i><sup><i>LacZ/d(neo)</i></sup> mutants. Mann-Whitney and Student-<i>t</i> test.</p

    Ectopic Met in limb mesenchyme alters HGF bioavailability.

    No full text
    <p>(A) Scheme illustrating the experimental procedure employed for evaluating through MDCK cell scattering the bioavailability of HGF from control and <i>Del-R26</i><sup><i>Met</i></sup> mutant limb mesenchymal cells or from dissected forelimbs. The scheme indicates the experimental procedure applied for collecting media conditioned by limb mesenchymal cells for biochemical analysis (top; shown in <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1005533#pgen.1005533.s009" target="_blank">S9A Fig</a>), for MDCK scattering assays using co-cultures with limb mesenchymal cells (middle; shown in Fig 9C, 9D and 9E) or with dissected limbs (bottom; shown in <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1005533#pgen.1005533.s009" target="_blank">S9B Fig</a>). (B) Pictures of MDCK colonies showing the three categories that were defined to determine the extent of cell contact and spreading for quantification studies of scattering response. (C) Quantitative analysis of MDCK cell scattering in co-cultures with control limb mesenchymal cells in the absence (no) and in the presence of the Met inhibitor PHA665752 (PHA; 1μM), cryzotinib (Cryzo; 1μM), or SU11274 (SU; 1μM). (D) Quantitative analysis of MDCK cell scattering in co-cultures with control limb mesenchymal cells in the absence (no) and in the presence of the anti-HGF blocking antibodies (anti-HGF; 30μg/ml). (E) Quantitative analysis of MDCK cell scattering in co-cultures with control or <i>Del-R26</i><sup><i>Met</i></sup> limb mesenchymal cells. Note a drastic reduction in the scattering response when MDCK cells are co-cultured with <i>Del-R26</i><sup><i>Met</i></sup> mutant cells (control: n = 4; <i>Del-R26</i><sup><i>Met</i></sup>: n = 3). Mann-Whitney and Student-<i>t</i> test.</p

    Myoblast migration is impaired in <i>Del-R26</i><sup><i>Met</i></sup> mutants.

    No full text
    <p>(A, B) Whole mount ISH of E10.5 embryos with <i>Lbx1</i> (A) and <i>Pax3</i> (B) probes showing drastic reduction of migrating myoblasts towards the developing tongue (arrowhead), fore and hind limbs. Bottom panel reports an enlargement at forelimb levels. (C, D) Quantification analyses of <i>Lbx1</i> (C) and <i>Pax3</i> (D) positive domains in forelimbs. Left panels: each plot represents the average signal distribution along the white line in forelimbs. Right panels: quantifications and statistical analyses of the sum of signal intensity based on intensity plots in left panels. Numbers of samples for <i>Lbx1</i>: control, n = 13; <i>Del-R26</i><sup><i>Met</i></sup>, n = 4; for <i>Pax3</i>: control, n = 11; <i>Del-R26</i><sup><i>Met</i></sup>, n = 8. The sum of <i>Pax3</i> signal intensity was calculated between point A and B: A indicating a fixed position between the somites and the limb whereas B being placed at a fixed distance from A. Note almost lack of signal in <i>Del-R26</i><sup><i>Met</i></sup> mutants. Scale: 500μm. Mann-Whitney and Student-<i>t</i> test.</p

    Enhanced Met expression levels in <i>Del-R26</i><sup><i>Met</i></sup> myoblasts does not perturb activation of downstream signalling effectors.

    No full text
    <p>Limb transverse sections of E10.5 control and <i>Del-R26</i><sup><i>Met</i></sup> embryos showing the distribution of phospho-Met (on Tyr<sub>1234–1235</sub>), phospho-Akt, phospho-ERKs (red) and of Pax3 protein (green) in myoblasts. Note ectopic phospho-Met in limb mesenchyme (arrowheads) and in non-migrating myoblasts (arrows) in <i>Del-R26</i><sup><i>Met</i></sup> mutants. Asterisks indicate non-specific staining in blood cells. Scale: 100μm.</p

    Triflorcas elicits a selective gene expression profile on stress and toxicity pathways.

    No full text
    <p>(A) The expression profile of 84 genes related to cell stress and toxicity was analyzed in GTL-16 cells. Cells were treated with either Triflorcas (black columns; 3 µM) or SU11274 (grey columns; 1 µM) for 24 hours, and gene expression was compared to that of untreated cells. Genes were grouped in clusters corresponding to oxidative/metabolic stress, heat shock, proliferation/carcinogenesis, growth arrest/senescence, inflammation, and apoptosis/necrosis signaling. Only statistically significant changes in gene expression are indicated (<i>P</i><0.05). Triflorcas altered the expression of only 14 genes compared to the alteration of 39 genes induced by SU11274 treatment. Notably, the expression of <i>cyp1A1</i> was increased 611-fold in the presence of Triflorcas. (B) Western blot analysis showing the up-regulation of CYP1A1 protein levels in cells exposed to Triflorcas (3 or 10 µM). Acacetin (ACA; 10 µM) treatment prevented CYP1A1 up-regulation by Triflorcas (TFC). (C) CYP1A1 up-regulation by Triflorcas also occurred in ErbB1-addicted cancer BT474 cells. Gefitinib (Gef; 10 µM) and SU11274 (SU; 2 µM) were used as controls.</p

    Triflorcas alters the phosphorylation status of cell cycle-related proteins.

    No full text
    <p>(A) The phosphorylation status of several cell cycle proteins was analyzed by applying the phospho-array KPSS 10.1 (Kinexus Bioinformatics). GTL-16 cells were treated with vehicle (control) or Triflorcas (TFC; 3 µM) for 72 hours (left and right panels, respectively). Red circles highlight constituents of the Akt/mTOR/S6 pathway. Green and blue circles surround nucleophosmin/B23 and Rb phospho-epitopes, respectively. (B) The graph shows the ratio of phosphorylation levels of the indicated proteins in cells untreated versus those exposed to Triflorcas. (C) Phosphorylation levels of nucleophosmin/B23 at Ser<sub>4</sub> and Rb at S<sub>780</sub> were increased and decreased, respectively, in GTL-16 cells exposed to Triflorcas (TFC; 3 µM for 24 hours).</p

    Small molecule kinase interaction map for Triflorcas.

    No full text
    <p>Compound was screened against a KINOME<i>scan</i> (<a href="http://www.kinomescan.com" target="_blank">http://www.kinomescan.com</a>) panel of 98 kinases. (A) The table indicates the kinases for which Triflorcas reduced more than 30% the binding constant. The ligand binding for each kinase (% of control condition) is indicated. (B) TREEspot image of the 98 kinases screened with the position of Triflorcas targets: Abl wild-type, Abl E255K and Abl T315I mutant forms, IKK-beta, JAK2, MKNK1, and ZAP70. The complete dataset is shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0046738#pone.0046738.s008" target="_blank">Table S2</a>. TK: tyrosine kinase; TKL: tyrosine kinase like; STE: STE kinase; CK1: cell kinase1; AGC: PKA, PKC, PKG kinases; CAMK: calcium calmodulin-regulated kinase; CMGC: CDK, MAP, GSK, CDK-like kinase.</p

    Triflorcas impairs in vivo tumor growth of cancer cells carrying oncogenic Met, without causing major side effects.

    No full text
    <p>(A) Evolution of the body weight in mice, treated intra-peritoneally with either vehicle or Triflorcas (TFC; 30 mg.kg<sup>−1</sup> every day) showed no significant differences. Body weight is expressed as weight evolution over the 21 day-treatment period. (B) The weight of heart, spleen, kidney, and liver was evaluated in mice daily injected with Triflorcas (30 mg.kg<sup>−1</sup>) or vehicle for 21 days. No significant differences were observed. Values are expressed as means ± s.e.m. <i>P</i>>0.05. (C and D) Triflorcas treatment (i.p. 30 and 60 mg.kg<sup>−1</sup> every other day) reduced tumor volume (C) and weight (D) in nude mice injected sub-cutaneously with H1437 cells. Crizotinib (50 mg.kg<sup>−1</sup> daily) did not impair tumor growth. Values are reported as boxplots and expressed as means ± s.e.m. *<i>P</i><0.05; Student-<i>t</i> test.</p
    corecore