von Willebrand factor directly interacts with DNA from neutrophil extracellular traps

S Grässle, V Huck, KI Pappelbaum… - … , and vascular biology, 2014 - Am Heart Assoc
S Grässle, V Huck, KI Pappelbaum, C Gorzelanny, C Aponte-Santamaría, C Baldauf
Arteriosclerosis, thrombosis, and vascular biology, 2014Am Heart Assoc
Objective—Inflammatory conditions provoke essential processes in the human vascular
system. It leads to the formation of ultralarge von Willebrand factor (VWF) fibers, which are
immobilized on the endothelial cell surface and transform to highly adhesive strings under
shear conditions. Furthermore, leukocytes release a meshwork of DNA (neutrophil
extracellular traps) during the process of the recently discovered cell death program
NETosis. In the present study, we characterized the interaction between VWF and DNA and …
Objective
Inflammatory conditions provoke essential processes in the human vascular system. It leads to the formation of ultralarge von Willebrand factor (VWF) fibers, which are immobilized on the endothelial cell surface and transform to highly adhesive strings under shear conditions. Furthermore, leukocytes release a meshwork of DNA (neutrophil extracellular traps) during the process of the recently discovered cell death program NETosis. In the present study, we characterized the interaction between VWF and DNA and possible binding sites to underline the role of VWF in thrombosis and inflammation besides its function in platelet adhesion.
Approach and Results
Both functionalized surfaces and intact cell layers of human umbilical vein endothelial cells were perfused with isolated, protein-free DNA or leukocytes from whole blood at distinct shear rates. DNA–VWF interaction was monitored using fluorescence microscopy, ELISA-based assays, molecular dynamics simulations, and electrostatic potential calculations. Isolated DNA, as well as DNA released by stimulated leukocytes, was able to bind to shear-activated, but not inactivated, VWF. However, DNA–VWF binding does not alter VWF degradation by a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13. Moreover, DNA–VWF interaction can be blocked using unfractionated and low-molecular-weight heparin, and DNA–VWF complexes attenuate platelet binding to VWF. These findings were supported using molecular dynamics simulations and electrostatic calculations of the A1- and A2-domains.
Conclusions
Our findings suggest that VWF directly binds and immobilizes extracellular DNA released from leukocytes. Therefore, we hypothesize that VWF might act as a linker for leukocyte adhesion to endothelial cells, supporting leukocyte extravasation and inflammation.
Am Heart Assoc