Abstract

Nitric oxide (NO) produced by endothelial cells in response to cytokines displays anti-inflammatory activity by preventing the adherence, migration and activation of neutrophils. The molecular mechanism by which NO operates at the blood-endothelium interface to exert anti-inflammatory properties is largely unknown. Here we show that on endothelial surfaces, NO is associated with the sulfhydryl-rich protein tissue transglutaminase (TG2), thereby endowing the membrane surfaces with anti-inflammatory properties. We find that tumor necrosis factor-α-stimulated neutrophil adherence is opposed by TG2 molecules that are bound to the endothelial surface. Alkylation of cysteine residues in TG2 or inhibition of endothelial NO synthesis renders the surface-bound TG2 inactive, whereas specific, high affinity binding of S-nitrosylated TG2 (SNO-TG2) to endothelial surfaces restores the anti-inflammatory properties of the endothelium, and reconstitutes the activity of endothelial-derived NO. We also show that SNO-TG2 is present in healthy tissues and that it forms on the membranes of shear-activated endothelial cells. Thus, the anti-inflammatory mechanism that prevents neutrophils from adhering to endothelial cells is identified with TG2 S-nitrosylation at the endothelial cell-blood interface.

Highlights

  • The physical interaction between neutrophils and activated endothelium is among the earliest events in inflammation[1]

  • Nitric oxide (NO) bioactivity can influence both the release of inflammatory cytokines and the expression of cell adhesion molecules, and it is recognized that production of NO by endothelial cells regulates neutrophil adhesion in vivo[6,7,8,9,10,11]

  • We sought a model system to examine the effect of surface-bound TG2 on polymorphic neutrophil (PMN) adherence and to test the influence of NO under inflammatory condition

Read more

Summary

Results

TG2 is bound constitutively to endothelial surfaces at the blood interface and transcriptionally up-regulated by TNFα17,31. GST-TG2/C277A binding increased following TNFα treatment of HUVECs. GST-TG2-treated samples showed multiple high molecular weight bands, likely reflecting crosslinking by canonical TG2 activity (Supplemental Fig. 3). Recombinant TG2/C277A was added to HUVEC monolayers after the activation with TNFα, and PMN adherence was monitored under physiological shear (to simulating blood flow). Following addition of TG2/C277A to HUVECs, activation of eNOS by shear stress resulted in the S-nitrosylation of about 14% of exogenous TG2/C277A thiols (comparable to results with aortic endothelial cells[18]). Shear stress activation of eNOS led to a large decrease in the adherence of PMNs, and this effect was abrogated by treatment with the NOS inhibitor L-NMMA, whereas addition of S-nitrosylated TG2/C277A inhibited PMN adherence in the presence of L-NMMA (Fig. 4). We show that GAPDH, a prototypic SNO-protein[32], is readily detected under these conditions

Discussion
Author Contributions
Additional Information
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