14.01.2013 Views

Visit our Expo - Redox and Inflammation signaling 2012

Visit our Expo - Redox and Inflammation signaling 2012

Visit our Expo - Redox and Inflammation signaling 2012

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 3<br />

Cell adhesion to the extracellular matrix increases the proliferation of acute myeloid<br />

leukemia (AML) cells <strong>and</strong> up-regulates the CDC25A phosphatase.<br />

Anne Fern<strong>and</strong>ez-Vidal, Loïc Ysebaert, Christine Didier, Stéphane Manenti.<br />

Centre de physiopathologie Toulouse-Purpan, INSERM U563-IFR30, Département<br />

« Oncogénèse et signalisation dans les cellules hématopoïétiques », CHU Purpan, 3 place<br />

du Dr Baylac 31024 Toulouse Cedex 3, France. E-mail : fervidal@toulouse.inserm.fr.<br />

Microenvironmental stimuli are key factors in normal <strong>and</strong> leukemic hematopoietic<br />

differenciation. In this contexte, we investigated the influence of the major extracellular<br />

matrix component, fibronectin, on the proliferative state of acute myeloid leukemia <strong>and</strong><br />

normal hematopoietic cells. Although fibronectin inhibited the proliferation rate of immature<br />

normal CD34+ cells, it stimulated the proliferation of immature leukemic cells at similar<br />

differentiation state (KG1a cell line). These data suggest that hematopoietic cells shift from a<br />

negative to a positive proliferative response to cell adhesion during the leukemogenic<br />

transformation process. As a general feature, we found that cell adhesion to fibronectin matrix<br />

increases the proliferation of various cell lines. Importantly, we identified the dual specificity<br />

phosphatase Cdc25A as a key effector of this effect. Indeed, adhesion to fibronectin of<br />

leukemic cells dramatically up-regulates the cellular level of this protein, in good correlation<br />

with the proliferative response. By contrast, Cdc25A protein down-regulation was observed in<br />

normal CD34+ cells, nicely fitting with the inhibition of the proliferation observed in these<br />

cells. Furthermore, siRNA- <strong>and</strong> pharmacological inhibitor-based experiments strongly suggest<br />

that Cdc25A regulation in this context is indeed involved in the adhesion –dependent<br />

proliferation process. Altogether, these data demonstrate for the first time an integrindependent<br />

Cdc25A protein regulation, making of this cell cycle effector an important link<br />

between extracellular stimuli <strong>and</strong> the cell cycle machinery. This also make of this phosphatase<br />

<strong>and</strong> of its molecular regulators, a potential therapeutic target in the acute myeloid leukemia<br />

pathologies.<br />

- 269 -

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!