Abstract

Integrin αIIbβ3 plays a central role in the adhesion and aggregation of platelets and thus is essential for hemostasis and thrombosis. Integrin activation requires the transmission of a signal from the small cytoplasmic tails of the α or β subunit to the large extracellular domains resulting in conformational changes of the extracellular domains to enable ligand binding. Hydrogen peroxide-inducible clone-5 (Hic-5), a member of the paxillin family, serves as a focal adhesion adaptor protein associated with αIIbβ3 at its cytoplasmic tails. Previous studies suggested Hic-5 as a novel regulator of integrin αIIbβ3 activation and platelet aggregation in mice. To assess this in more detail, we generated Hic-5-null mice and analyzed activation and aggregation of their platelets in vitro and in vivo. Surprisingly, lack of Hic-5 had no detectable effect on platelet integrin activation and function in vitro and in vivo under all tested conditions. These results indicate that Hic-5 is dispensable for integrin αIIbβ3 activation and consequently for arterial thrombosis and hemostasis in mice.

Highlights

  • Vessel wall injury results in the exposure of the subendothelial extracellular matrix which initiates stable platelet adhesion and aggregation [1,2]. These processes are crucial for normal hemostasis, but in diseased vessels they may lead to pathological thrombus formation and infarction of vital organs [3]

  • Platelet adhesion and aggregation are mediated by integrins, heterodimeric transmembrane receptors composed of α and β subunits that are expressed in a low affinity state under resting conditions

  • We used embryonic stem cells provided by CSD, a collaborative team at the Children's Hospital Oakland Research Institute, and the University of California at Davis, within the Knockout Mouse Program (KOMP) with a knockout first approach targeting exon 1 of the Tgfb1i1 gene leading to efficient loss of Hydrogen peroxide-inducible clone-5 (Hic-5) protein in Tgfb1i1+/- mice

Read more

Summary

Introduction

Vessel wall injury results in the exposure of the subendothelial extracellular matrix which initiates stable platelet adhesion and aggregation [1,2]. We generated Hic-5-null (Tgfb1i1-/-) mice and found, in stark contrast to the study by KimKaneyama et al [14], unaltered platelet integrin function in vitro and in vivo. For adhesion to vWF, cover slips were coated with rabbit anti-human vWF antibody at 4°C o/n, washed with PBS, blocked for 1 h with 1% BSA in H2O and incubated with 100 μl murine serum obtained from control mice.

Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call