Abstract

Proteinase 3 (PR3), the autoantigen in granulomatosis with polyangiitis, is expressed at the plasma membrane of resting neutrophils, and this membrane expression increases during both activation and apoptosis. Using surface plasmon resonance and protein-lipid overlay assays, this study demonstrates that PR3 is a phosphatidylserine-binding protein and this interaction is dependent on the hydrophobic patch responsible for membrane anchorage. Molecular simulations suggest that PR3 interacts with phosphatidylserine via a small number of amino acids, which engage in long lasting interactions with the lipid heads. As phosphatidylserine is a major component of microvesicles (MVs), this study also examined the consequences of this interaction on MV production and function. PR3-expressing cells produced significantly fewer MVs during both activation and apoptosis, and this reduction was dependent on the ability of PR3 to associate with the membrane as mutating the hydrophobic patch restored MV production. Functionally, activation-evoked MVs from PR3-expressing cells induced a significantly larger respiratory burst in human neutrophils compared with control MVs. Conversely, MVs generated during apoptosis inhibited the basal respiratory burst in human neutrophils, and those generated from PR3-expressing cells hampered this inhibition. Given that membrane expression of PR3 is increased in patients with granulomatosis with polyangiitis, MVs generated from neutrophils expressing membrane PR3 may potentiate oxidative damage of endothelial cells and promote the systemic inflammation observed in this disease.

Highlights

  • Proteinase 3 Affects Microvesicle Production and Function eat me signal, membrane PR3 can perpetuate inflammation by increasing the production of proinflammatory cytokines including TNF␣, MCP1, and IL-6 [12]

  • Data in A, C, and D are represented as respiratory burst generated from 1 ϫ 106 MVs; n ϭ 7 independent MV preparations used on neutrophils isolated from different donors (*/#, p Ͻ 0.05; **/##, p Ͻ 0.01; one-way analysis of variance (ANOVA))

  • Data in B represent percent change compared with pcDNA obtained from n ϭ 5 independent MV preparations used on neutrophils isolated from different donors (**, p Ͻ 0.01; one-way ANOVA)

Read more

Summary

Affects the Production and Function of Microvesicles*

Anti-neutrophil cytoplasmic antibodies (ANCAs) directed against either PR3 and myeloperoxidase, the autoantigens for GPA and microscopic polyangiitis, respectively, stimulated the production of MVs from human neutrophils primed with TNF␣ [20] These MVs were able to bind to endothelial cells to induce expression of proinflammatory mediators including IL-6 and IL-8 and upregulate ICAM1, an adhesion molecule involved in facilitating leukocyte endothelial transmigration [20]. Treatment of human umbilical vein endothelial cells with neutrophil-derived MVs resulted in a loss of membrane integrity and morphological changes characteristic of cell injury [21] Based on these studies, it seems likely that MVs generated during autoimmune vasculitis do play an active role in promoting inflammation and endothelial cell damage. Given that PR3 displays a high affinity for lipids, we investigated whether this protein could bind to PS and whether this interaction affected microvesicle-dependent biological processes known to modulate inflammation

Experimental Procedures
Results
Discussion
Full Text
Published version (Free)

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