Primary megakaryocytes reveal a role for transcription factor NF-E2 in integrin αIIbβ3 signaling

M Shiraga, A Ritchie, S Aidoudi, V Baron… - The Journal of cell …, 1999 - rupress.org
M Shiraga, A Ritchie, S Aidoudi, V Baron, D Wilcox, G White, B Ybarrondo, G Murphy
The Journal of cell biology, 1999rupress.org
Platelet integrin αIIbβ3 responds to intracellular signals by binding fibrinogen and triggering
cytoskeletal reorganization, but the mechanisms of αIIbβ3 signaling remain poorly
understood. To better understand this process, we established conditions to study αIIbβ3
signaling in primary murine megakaryocytes. Unlike platelets, these platelet precursors are
amenable to genetic manipulation. Cytokine-stimulated bone marrow cultures produced
three arbitrary populations of αIIbβ3-expressing cells with increasing size and DNA ploidy …
Platelet integrin αIIbβ3 responds to intracellular signals by binding fibrinogen and triggering cytoskeletal reorganization, but the mechanisms of αIIbβ3 signaling remain poorly understood. To better understand this process, we established conditions to study αIIbβ3 signaling in primary murine megakaryocytes. Unlike platelets, these platelet precursors are amenable to genetic manipulation. Cytokine-stimulated bone marrow cultures produced three arbitrary populations of αIIbβ3-expressing cells with increasing size and DNA ploidy: small progenitors, intermediate-size young megakaryocytes, and large mature megakaryocytes. A majority of the large megakaryocytes bound fibrinogen in response to agonists, while almost none of the smaller cells did. Fibrinogen binding to large megakaryocytes was inhibited by Sindbis virus-mediated expression of isolated β3 integrin cytoplasmic tails. Strikingly, large megakaryocytes from mice deficient in the transcription factor NF-E2 failed to bind fibrinogen in response to agonists, despite normal surface expression of αIIbβ3. Furthermore, while megakaryocytes from wild-type mice spread on immobilized fibrinogen and exhibited filopodia, lamellipodia and Rho-dependent focal adhesions and stress fibers, NF-E2–deficient megakaryocytes adhered poorly. These studies establish that agonist-induced activation of αIIbβ3 is controlled by NF-E2–regulated signaling pathways that mature late in megakaryocyte development and converge at the β3 cytoplasmic tail. Megakaryocytes provide a physiologically relevant and tractable system for analysis of bidirectional αIIbβ3 signaling.
rupress.org